free article

12
Mesenchymal stem cell transplantation attenuates brain injury after neonatal stroke Cindy T.J. van Velthoven, PhD 1 , R. Ann Sheldon, BS 2 , Annemieke Kavelaars, PhD 3 , Nikita Derugin, MA 2 , Zinaida S. Vexler, PhD 2 , Hanneke L.D.M. Willemen, MSc 1 , Mirjam C.T. Maas, BS 1 , Cobi J. Heijnen, PhD 1,3 , and Donna M. Ferriero, MD 2 1 Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht, the Netherlands 2 Departments of Neurology and Pediatrics, University of California San Francisco, San Francisco, CA, USA 3 Department of Symptom Research, University of Texas, MD Anderson Cancer Centre, Houston, TX, USA Abstract Background and Purpose—Brain injury caused by stroke is a frequent cause of perinatal morbidity and mortality with limited therapeutic options. Mesenchymal stem cells (MSC) have been shown to improve outcome after neonatal hypoxic-ischemic brain injury mainly by secretion of growth factors stimulating repair processes. We investigated whether MSC-treatment improves recovery after neonatal stroke and whether MSC overexpressing brain-derived neurotrophic factor (MSC-BDNF) further enhances recovery. Methods—We performed 1.5-hour transient middle cerebral artery occlusion (MCAO) in 10- day-old rats. Three days after reperfusion, pups with evidence of injury by diffusion-weighted MRI were treated intranasally with MSC, MSC-BDNF or vehicle. To determine the effect of MSC-treatment, brain damage, sensorimotor function and cerebral cell proliferation were analysed. Results—Intranasal delivery of MSC- and MSC-BDNF significantly reduced infarct size and grey matter loss in comparison to vehicle-treated rats without any significant difference between MSC- and MSC-BDNF-treatment. Treatment with MSC-BDNF significantly reduced white matter loss with no significant difference between MSC- and MSC-BDNF-treatment. Motor deficits were also improved by MSC-treatment when compared to vehicle treated rats. MSC-BDNF-treatment resulted in an additional significant improvement of motor deficits 14 days post-MCAO, but there was no significant difference between MSC or MSC-BDNF 28 days post-MCAO. Furthermore, treatment with either MSC or MSC-BDNF induced long lasting cell proliferation in the ischemic hemisphere. Conclusions—Intranasal administration of MSC after neonatal stroke is a promising therapy for treatment of neonatal stroke. In this experimental paradigm MSC and BNDF-hypersecreting-MSC are equally effective in reducing ischemic brain damage. Corresponding author: Cindy T.J. van Velthoven, Ph.D., Room KC03.068.0, University Medical Center Utrecht, Lundlaan 6, 3584EA Utrecht, the Netherlands, Phone: +31 887554354, Fax: +31 887555311, [email protected]. Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. NIH Public Access Author Manuscript Stroke. Author manuscript; available in PMC 2014 May 01. Published in final edited form as: Stroke. 2013 May ; 44(5): 1426–1432. doi:10.1161/STROKEAHA.111.000326. NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

Upload: vuongthuan

Post on 31-Dec-2016

220 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: free article

Mesenchymal stem cell transplantation attenuates brain injuryafter neonatal stroke

Cindy T.J. van Velthoven, PhD1, R. Ann Sheldon, BS2, Annemieke Kavelaars, PhD3, NikitaDerugin, MA2, Zinaida S. Vexler, PhD2, Hanneke L.D.M. Willemen, MSc1, Mirjam C.T. Maas,BS1, Cobi J. Heijnen, PhD1,3, and Donna M. Ferriero, MD2

1Laboratory of Neuroimmunology and Developmental Origins of Disease, University MedicalCenter Utrecht, Utrecht, the Netherlands 2Departments of Neurology and Pediatrics, University ofCalifornia San Francisco, San Francisco, CA, USA 3Department of Symptom Research,University of Texas, MD Anderson Cancer Centre, Houston, TX, USA

AbstractBackground and Purpose—Brain injury caused by stroke is a frequent cause of perinatalmorbidity and mortality with limited therapeutic options. Mesenchymal stem cells (MSC) havebeen shown to improve outcome after neonatal hypoxic-ischemic brain injury mainly by secretionof growth factors stimulating repair processes. We investigated whether MSC-treatment improvesrecovery after neonatal stroke and whether MSC overexpressing brain-derived neurotrophic factor(MSC-BDNF) further enhances recovery.

Methods—We performed 1.5-hour transient middle cerebral artery occlusion (MCAO) in 10-day-old rats. Three days after reperfusion, pups with evidence of injury by diffusion-weightedMRI were treated intranasally with MSC, MSC-BDNF or vehicle. To determine the effect ofMSC-treatment, brain damage, sensorimotor function and cerebral cell proliferation wereanalysed.

Results—Intranasal delivery of MSC- and MSC-BDNF significantly reduced infarct size andgrey matter loss in comparison to vehicle-treated rats without any significant difference betweenMSC- and MSC-BDNF-treatment. Treatment with MSC-BDNF significantly reduced white matterloss with no significant difference between MSC- and MSC-BDNF-treatment. Motor deficits werealso improved by MSC-treatment when compared to vehicle treated rats. MSC-BDNF-treatmentresulted in an additional significant improvement of motor deficits 14 days post-MCAO, but therewas no significant difference between MSC or MSC-BDNF 28 days post-MCAO. Furthermore,treatment with either MSC or MSC-BDNF induced long lasting cell proliferation in the ischemichemisphere.

Conclusions—Intranasal administration of MSC after neonatal stroke is a promising therapy fortreatment of neonatal stroke. In this experimental paradigm MSC and BNDF-hypersecreting-MSCare equally effective in reducing ischemic brain damage.

Corresponding author: Cindy T.J. van Velthoven, Ph.D., Room KC03.068.0, University Medical Center Utrecht, Lundlaan 6,3584EA Utrecht, the Netherlands, Phone: +31 887554354, Fax: +31 887555311, [email protected].

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to ourcustomers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review ofthe resulting proof before it is published in its final citable form. Please note that during the production process errors may bediscovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

NIH Public AccessAuthor ManuscriptStroke. Author manuscript; available in PMC 2014 May 01.

Published in final edited form as:Stroke. 2013 May ; 44(5): 1426–1432. doi:10.1161/STROKEAHA.111.000326.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 2: free article

Keywordsneonatal stroke; cerebral ischemia; postnatal; cell transplantation; mesenchymal stem cells

IntroductionNeonatal stroke occurs in approximately 1 in 4,000 live births and is associated withsignificant morbidity and mortality.1 Neonates suffering from perinatal stroke often developlong term disabilities including motor deficits, cognitive dysfunction and epilepsy.2

Currently there are no accepted treatment options for this vulnerable group of infants.Therefore, development of new treatment strategies is urgently needed.

Over the past decades a large number of studies have evaluated potential therapies toprevent progression of injury via pharmacological neuroprotection. More recently, means toenhance repair of the damaged immature brain are being investigated. Several studies, usingdifferent types of brain injury including adult stroke models of middle cerebral arteryocclusion (MCAO) and neonatal hypoxic-ischemic brain injury, have shown thatadministration of MSC promotes functional neurological recovery.3,4 This beneficial effectof MSC-transplantation might involve replacement of damaged cells by the transplantedcells. However, data in the literature suggest that it is more likely that transplanted MSCinduce repair by stimulating secretion of growth and differentiation factors therebyproviding an environment that stimulates repair processes like neurogenesis andangiogenesis.5 We and others have shown that in response to the growth factor environmentin the damaged brain, MSC secrete several factors which have the potential to stimulaterepair processes in the brain.5–7

In this study we investigated whether MSC transplantation has beneficial effects onfunctional outcome and lesion volume in a rat model of neonatal stroke. Furthermore, wedetermined if the therapeutic potential of MSC could be enhanced by genetically modifyingthe MSC to secrete more brain-derived neurotrophic factor (BDNF).

MethodsAdenoviral vector

Adenoviral vector (pAd-HM41-K7; Alphagen) carrying the gene for polylysine-mutatedfiberknot was constructed as described previously.8 Mouse brain-derived neurotrophic factor(BDNF) cDNA was cloned using RT-PCR with total RNA isolated from brain as template.BDNF sequence was confirmed by sequencing and comparison to GenBank sequenceNM_007540. Mouse BDNF-primer sequences were Forward 5’-TCTAGACACCCCACCATGACCATCCTTTTCCTT-3’, Reverse 5’-TCTTCCCCTTTTAATGGTCAGT-3’. BDNF cDNA was coupled to an IRES-eGFPsequence to allow labeling of infected cells. The BDNF-IRES-eGFP sequence was insertedinto the pShuttle2 vector between the XbaI and AflII sites resulting in the pShuttle2-BDNF-IE plasmid. pAd-HM41-K7-BDNF-IE was constructed by ligation of I-CeuI/PI-SceI-digested pShuttle2-BDNF-IE with I-CeuI/PI-SceI-digested pAd-HM41-K7. An emptyvector control was generated and consisted only of the IRES-eGFP sequence insert (pAd-HM410K7-IE).

Virus particles were generated by transfection of PacI-digested pAd-HM41-K7-BDNF-IEinto 293 cells with Lipofectamine2000 (Invitrogen). Before being used, virus titer wasdetermined and stocks were examined for potential contamination with replication-competent viruses.

van Velthoven et al. Page 2

Stroke. Author manuscript; available in PMC 2014 May 01.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 3: free article

MSCRat Sprague-Dawley MSC (GIBCO) were cultured according to manufacturer’s instructions.Cells were negative for myeloid and haematopoietic cell lineage specific antigens andpositive for CD29, CD44, CD90 and CD106. MSC were seeded at a density of 3×106 cellsper 25cm2 flask, exposed to pAd-HM41-K7-BDNF-IE or pAd-HM41-K7-IE virus particlesin 7.5ml DMEM for 6 hours after which cultures were washed 3 times with DMEM and re-cultured with normal medium. Following infection, MSC were cultured for an additional 24hours after which transplantation was performed.

AnimalsAll animal research was approved by the University of California San Francisco InstitutionalAnimal Care and Use Committee and performed in accordance with the Guide for Care andUse of Laboratory Animals (U.S. Department of Health and Human Services, PublicationNumber 85-23, 1985).

Transient 1.5-hour right MCAO was performed in 10d-old Sprague-Dawley rats asdescribed previously.9 Briefly, surgery was performed on spontaneously breathing pupsanesthetized with 1.75% isoflurane in a mixture of 70% N2O and 30% O2. The internalcarotid artery (ICA) was dissected and a temporary ligature was applied at its origin using a6-0 silk suture. A second suture was looped around the ICA, just above the pterygopalatineartery, and retracted laterally to prevent retrograde blood flow. A small arteriotomy wasmade in the proximal isolated ICA segment, a coated 6-0 Dermalon filament was insertedand advanced 7.5–8.5mm depending on the animal’s weight and secured with a temporarysuture. The filament and both sutures around the ICA were removed 1.5 hours later,reestablishing blood flow.

Spin-echo planar diffusion-weighted MRI (DWI) was performed using a 7T magnet 3 daysafter MCAO to identify injured animals; only animals with injury extending throughout themiddle cerebral artery territory on DWI were used. Injury volume was determined in 6consecutive 2mm thick coronal sections. A total of 36 pups met inclusion criteria based onDWI after MCAO (85%). Overall survival of injured animals was 87%.

At 3 days post-MCAO, MSC, BDNF-secreting-MSC (MSC-BDNF) or vehicle wasdelivered intranasally to awake rats. 30 Minutes prior to MSC or vehicle administration twodoses of 5µl hyaluronidase (Sigma-Aldrich) in PBS were applied to each nostril andspontaneously inhaled.10 Subsequently, a total of 1×106 MSC in 20µl PBS or vehicle wereadministered as two doses of 5µl applied to each nostril.

To evaluate cell proliferation/survival, rats received ethynyldeoxyuridine (EdU; 50mg/kg,i.p.; Invitrogen) at days 3 to 5 post-MCAO and bromodeoxyuridine (BrdU; 50mg/kg, i.p.;Sigma-Aldrich) at days 21 to 23 post-MCAO. Animals were sacrificed at 28 days post-MCAO and perfused with 4% paraformaldehyde in PBS.

Functional outcomeThe cylinder-rearing test was used to assess forelimb use asymmetry.11 The weight-bearingforepaw(s) to contact the wall during a full rear was recorded as left (impaired), right (non-impaired) or both. Paw preference was calculated as [(nonimpaired–impaired)/(nonimpairedforepaw+impaired forepaw+both)]×100. Adhesive removal test was performed at 28 daysafter MCAO. Stickers (tough-spots, Diversified Biotech, Boston MA) were placed on leftand right forepaw and the latency to removal was recorded. The mean time until completeremoval of three stickers per forepaw was recorded. Sticker placement on left and rightforepaw was alternated between and within animals.11

van Velthoven et al. Page 3

Stroke. Author manuscript; available in PMC 2014 May 01.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 4: free article

Histology and ImmunohistochemistryCoronal paraffin sections (10µm) were incubated with mouse-anti-MBP (SternbergerMonoclonals) or mouse-anti-MAP2 (Sigma-Aldrich) and binding was visualized with aVectastain ABC kit (Vector Laboratories). Volumetric injury analysis was performed on aseries of six MAP2, MBP and cresyl violet stained sections (ImageJ, NIH). Injury volumewas expressed as ratio ipsi- to contralateral hemisphere volume.4,12

For cell proliferation analysis, sections were incubated with biotinylated sheep-anti-BrdUand rabbit-anti-Ki67 (Abcam). Visualization was done with AlexaFluor-594 conjugatedstreptavidin and donkey-anti-rabbit AlexaFluor488 (Molecular Probes). EdU incorporationwas detected by incubating sections in 100mM Tris containing 0.5mM CuSO4, 50mMascorbic acid and 10µM AlexaFluor-594-azide (Molecular Probes). EdU- and BrdU-positivecells were counted in three high magnification fields in the striatum of the damagedhemisphere in three sections per brain. Ki67-positive cells were counted in SVZ in threesections per brain.

Statistical analysisAll data are expressed as mean±SEM. Functional outcome measured with cylinder rearingtest and adhesive removal test were analysed using two-way ANOVA with Fisher’s LSDpost-tests. Histological measures were analyzed using one-way ANOVA with Bonferronipost-tests. p<0.05 was considered statistically significant.

ResultsMSC treatment following neonatal stroke reduces lesion volume

Rats underwent MCAO at p10 and were treated with MSC, MSC-BDNF or vehicle at 3 daysafter the insult. Animals that underwent MCAO had significant brain tissue loss in theipsilateral hemisphere at 28 days post-MCAO (Fig. 1A). Following treatment with MSCthere was a significant reduction in ipsilateral tissue loss in comparison to vehicle-treatedrats (p<0.05). Treatment with MSC-BDNF resulted in a reduction of tissue loss whencompared to (p<0.01 vs. vehicle), but not significantly different from MSC-treated rats.

We also analyzed the effect of MCAO and subsequent MSC treatment on grey and whitematter loss. At 28 days post-MCAO MAP2-positive area loss, as a measure of grey matterinjury, was significantly lower after treatment with MSC or MSC-BDNF in comparison tovehicle treated rats (p<0.05 and p<0.01 respectively). Treatment with MSC-BDNF did notresult in significantly lower MAP-2 loss than treatment with MSC (Fig. 1B).

Injury to white matter was analyzed by measuring MBP-positive area. MCAO resulted insignificant loss of MBP-positive tissue in vehicle-treated rats (Fig. 1C). Treatment withMSC did not significantly decrease MBP area loss when compared to vehicle-treated rats.Treatment with MSC-BDNF, however, did cause a significant reduction in MBP-positivearea loss (p<0.05) with no significant difference between MSC and MSC-BDNF treatment.

MSC transplantation after neonatal stroke reduces motor deficitsTo determine the extent of lateralizing motor deficits caused by our stroke model, thecylinder rearing test was performed at 14, 21 and 28 days after MCAO. In this test, sham-operated rats did not show any paw preference (Fig. 2A). MCAO caused lateralizationshown by a ~30% preference to use the right unimpaired forepaw in vehicle-treated rats.Following treatment with MSC, performance in the cylinder rearing test significantlyimproved at all three time points measured in comparison to vehicle-treated rats. At 14 dayspost-MCAO forepaw impairment in MSC-BDNF treated rats was significantly lower than in

van Velthoven et al. Page 4

Stroke. Author manuscript; available in PMC 2014 May 01.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 5: free article

MSC-treated rats (p<0.05). However, the effect of MSC-BDNF-treatment was onlytemporary as there was no difference between MSC-BDNF and MSC-treated rats at 28 dayspost-MCAO.

At 28 days post-MCAO all rats were subjected to the adhesive removal test. This testevaluates sensory and motor deficits related to the paw. Sham-operated controls did notshow a difference in adhesive removal latency between left and right forepaw (Fig. 2B).After MCAO all rats were impaired in the adhesive removal test meaning that the latency toremove the sticker from the impaired forepaw was significantly higher than removing thesticker from the unimpaired forepaw. More importantly, MSC- and MSC-BDNF-treated ratsshowed a reduced latency to remove the adhesive from the impaired forepaw whencompared to vehicle treated rats. There was no difference in adhesive removal latencybetween rats treated with MSC or MSC-BDNF.

MSC treatment following neonatal stroke induces long lasting cell proliferation in theipsilesional striatum and subventricular zone (SVZ)

To determine the effect of MSC transplantation and modified-MSC transplantation on cellproliferation in the brain after neonatal stroke, rats were injected with cell proliferationmarkers EdU on days 3 to 5 post-MCAO and, to check for late proliferation, with BrdU ondays 21 to 23 post-MCAO. At 28 days post-MCAO the number of EdU- and BrdU-labelledcells and expression of the cell proliferation marker Ki67 were analysed.

At 28 days after HI only some EdU- and BrdU-positive cells could be detected in thesubventricular zone. However, in the injured striatum and near the ischemic boundary zone asignificant number of both EdU- and BrdU-positive cells were visible. No increase in EdU-positive cell number in the striatum was detected at 28 days post-MCAO in vehicle-treatedrats in comparison to sham-operated controls. Treatment with either MSC or MSC-BDNFsignificantly increased EdU-positive cells number in the ipsilateral striatum. There was nodifference in the number of EdU-positive cells between MSC- and MSC-BDNF treated rats(Fig. 3A and D), indicating that there was no difference in cell proliferation induced byeither treatment with MSC or MSC-BDNF.

Measurement of late cell proliferation by analysis of BrdU-positive cell number, showedthat MCAO with vehicle-treatment had no effect on cell proliferation at 21–23 days postMCAO. Following treatment with MSC and MSC-BDNF the number of BrdU-positive cellsincreased when compared to vehicle treated rats. However, there was no additional effect ofMSC-BDNF when compared to MSC treatment (Fig. 3B and D).

At 28 days post-MCAO cell proliferation in the MSC-treated rats, proliferation was stillincreased as compared to vehicle-treated MCAO rats as witnessed by increased Ki67-positive cell number in the subventricular zone. Treatment with MSC-BDNF didsignificantly increase the number of Ki67-positive cells in the SVZ when compared tovehicle treated rats, but was not different from treatment with MSC alone (Fig. 3C and D).

DiscussionThis study, for the first time, shows that intranasal application of MSC or BDNFhypersecreting MSC following neonatal stroke in rats effectively reduces long-termfunctional impairment and infarct volume and increases cell proliferation in the ischemichemisphere. Improved outcome after transplantation of MSC has been attributed toinhibition of inflammation, neuroprotection, direct replacement of lost tissue by MSC andstimulation of endogenous repair processes.13,14 MSCs secrete a variety of factors, includingneurotrophins like BDNF and NGF, growth factors like VEGF and IGF and interleukins.6,7

van Velthoven et al. Page 5

Stroke. Author manuscript; available in PMC 2014 May 01.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 6: free article

Recently Kim et al.15 showed that intraventricular administration of human umbilical cordblood-derived MSC 6 hours after onset of neonatal stroke in a non-reperfusion modelreduced ipsilesional tissue loss. This was accompanied by a dampened inflammatoryresponse and decreased cell death. It has been suggested that the beneficial effects of MSCare mediated by the immuno-modulatory effect of MSC as well as by delivery of trophicfactors.13,14 We observed a decrease in lesion volume after MSC treatment. However, iInthe adult MCAO model, treatment with MSC has been shown to improve neurologicalfunction without reducing lesion volume.13,16 All of these results could either be interpretedas MSC-induced endogenous repair or MSC-induced neuroprotection. In some studies injuvenile animals there is evidence for significant late neuronal loss.17 However, we did notobserve a deterioration of neuronal function between day 3 and 10 post-HI when using thehypoxia-ischemia model in p9 mice.4 Therefore we proposed that MSC transplantation inthe neonatal hypoxia-ischemia model may well promote endogenous neuronal repair. In themodel of neonatal stroke currently described we do not know at present whether lateneuronal loss develops, so we cannot conclude whether MSC mainly function by protectingthe brain against late neurodegeneration and/or by endogenous repair mechanisms. The factthat treatment with MSC increase cell proliferation in the SVZ lasting for at least 28 dayspost-MCAO may indicate that endogenous repair processes are contributing to thetherapeutic effects of MSC in our neonatal stroke model.

Recent results from our group showed that MSC-transplantation 10 days after induction ofneonatal HI induces various changes in the cytokine and growth factor environment.5,7

Following transplantation of MSC after neonatal HI the gene expression profile in the brainshifts towards a growth promoting environment. In vivo this is visible as increased cellproliferation, formation of new neurons and decreased gliosis suggesting decreasedinflammation following MSC treatment of ischemic brain injury.4 In the present study,transplantation of both MSC and MSC-BDNF resulted in long-lasting cell proliferation inthe SVZ. The SVZ contains neural stem cells, which under normal circumstancesdifferentiate into neurons and migrate towards the olfactory bulb.18 Local injury canstimulate cell proliferation in the SVZ upon which cells migrate towards the injured area anddifferentiate into neurons, oligodendrocytes and asrocytes.19 Our results in this study showthat MSC treatment increases cell proliferation for at least 28 days post-MCAO. Cellproliferation markers EdU and BrdU were injected, directly after administration of the MSCand 2 weeks later respectively, to determine cell proliferation during the course of theexperiment. At 28 days after HI only some EdU- and BrdU-positive cells could be detectedin the SVZ. However, in the injured striatum and near the ischemic boundary zone asignificant number of both EdU- and BrdU-positive cells was visible. These EdU- or BrdU-labelled cells probably originate from the subventricular zone and migrated towards theinjured area where they potentially contribute to repair of the injured tissue and decreasedlesion volume that was observed after MSC transplantation.

Administration of MSC following neonatal stroke significantly reduces lateralizing motordeficits. Besides the effect of MSC-transplantation following neonatal stroke on motor andsomatosensory function, it is likely that also cognitive function is improved in rats followingtreatment with MSC. Recent studies have shown that neurorestorative treatments like MSC-transplantation or erythropoietin can indeed improve cognitive function following ischemicbrain damage.20,21 A study on cognitive effects may be of importance in view of repair ofwhite matter structures induced by treatment with MSC or MSC-BDNF (see Fig 2).

Neurotrophic factors play an important role in repair of ischemic brain damage. BDNF, inparticular, is an important neurotrophic factor promoting neurogenesis and angiogenesis, itoffers neuroprotection, modulates inflammation and can improve synaptic plasticity after

van Velthoven et al. Page 6

Stroke. Author manuscript; available in PMC 2014 May 01.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 7: free article

ischemic brain injury.22–24 Furthermore, it has been shown that intracranial infusion ofBDNF using an osmotic pup can reduce infarct volume after stroke.

Our present study shows that the effect of MSC-BDNF administration seems to betemporary. Treatment with MSC-BDNF causes less lateralizing motor deficits measured atone week after administration of the stem cells after which motor deficits get morepronounced and at 4 weeks reach the level of rats treated with MSC alone. However, at 4weeks post-stroke BDNF-MSC are not more effective in reducing gross infarct size, greyand white matter loss and induction of cell proliferation than treatment with MSC alone. It ispossible that the additive effect of BDNF on transplantation of MSC transplantation of MSCobserved in the early measurement in the cylinder rearing test (day 14) is a direct effect ofBDNF on neuronal activity. BDNF plays a role in several stages of the development ofneural circuits, including neural stem cell survival and differentiation, axonal growth andrefinement of developing circuits.25 The effect of BDNF-MSC might be targeted towardsthe refinement of neural circuits that are developing. Addition of BDNF in this period ofdevelopment could lead to selective stabilization of some connections and elimination ofothers more effectively than MSC alone does.

In this study we used a non-integrating adenoviral construct to overexpress BDNF in MSC,meaning that BDNF expression subsides over time as the vector is removed from the cell.The temporary expression of BDNF might have been too short to induce long lasting effects.Therefore additional administrations of BDNF secreting MSC at later time points may benecessary to produce a more sustained effect. However, there is a time window ofadministration for MSC to be effective. We have recently shown, using the murine hypoxia-ischemia model, that a second dose of MSC at 17 days post-HI has no additive effect on topof a first dose at 10 days post-HI while two doses at 3 and 10 days post-HI effectivelyreduce HI brain damage.20

Apart from the fact that efficacy of (modified) MSC transplantation is of great importance,safety is a key issue in light of future clinical application of transplantation of (modified)MSC to babies with brain damage.26 Allogeneic MSC have been used for many years nowin the treatment of hematopoietic diseases which means that protocols regarding isolation,administration and safety are already established. The use of an adenovirus to modify MSCinstead of lenti- or retrovirus reduces the risk of developing neoplasms, since adenovirusesdo not integrate into the host genome. However, fate of transplanted cells and possibleadverse behavioural effects must be monitored closely in a phase II trial applying (modified)MSC transplantation in newborn babies with brain damage.

In summary, this study shows that intranasal application of BDNF-secreting MSC is equallyeffective in reducing grey and white matter loss and motor deficits and inducing cellproliferation after neonatal MCAO as use of MSC. Thus intranasal treatment with MSC maybe a promising therapy for neonatal ischemic brain damage.

The signals responsible for orchestrating repair processes such as neurogenesis, gliogenesisand angiogenesis are complex and depend on an intricate balance of various intra- andextracellular molecules.27 It may well be possible that MSC are equipped well enough toreact to the needs of the ischemic environment to stimulate repair processes in the neonatalischemic brain.

AcknowledgmentsSources of Funding

van Velthoven et al. Page 7

Stroke. Author manuscript; available in PMC 2014 May 01.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 8: free article

This study was funded by the European Union (HEALTH-F2-2009-241778, NEUROBID); Zon-MW Project (no116002003) and NIH (grant NS35902)

References1. Ferriero DM. Neonatal brain injury. N Engl J Med. 2004; 351:1985–1995. [PubMed: 15525724]

2. Kirton A, deVeber G. Advances in perinatal ischemic stroke. Pediatr Neurol. 2009; 40:205–214.[PubMed: 19218034]

3. Chen J, Li Y, Wang L, Lu M, Zhang X, Chopp M. Therapeutic benefit of intracerebraltransplantation of bone marrow stromal cells after cerebral ischemia in rats. J Neurol Sci. 2001;189:49–57. [PubMed: 11535233]

4. van Velthoven CT, Kavelaars A, van Bel F, Heijnen CJ. Mesenchymal stem cell treatment afterneonatal hypoxic-ischemic brain injury improves behavioral outcome and induces neuronal andoligodendrocyte regeneration. Brain Behav Immun. 2010; 24:387–393. [PubMed: 19883750]

5. van Velthoven CT, Kavelaars A, van Bel F, Heijnen CJ. Mesenchymal stem cell transplantationchanges the gene expression profile of the neonatal ischemic brain. Brain Behav Immun. 2011;25:1342–1348. [PubMed: 21473911]

6. Qu R, Li Y, Gao Q, Shen L, Zhang J, Liu Z, et al. Neurotrophic and growth factor gene expressionprofiling of mouse bone marrow stromal cells induced by ischemic brain extracts. Neuropathology.2007; 27:355–363. [PubMed: 17899689]

7. van Velthoven CT, Kavelaars A, van Bel F, Heijnen CJ. Repeated mesenchymal stem cell treatmentafter neonatal hypoxia-ischemia has distinct effects on formation and maturation of new neuronsand oligodendrocytes leading to restoration of damage, corticospinal motor tract activity, andsensorimotor function. J Neurosci. 2010; 30:9603–9611. [PubMed: 20631189]

8. Mizuguchi H, Sasaki T, Kawabata K, Sakurai F, Hayakawa T. Fiber-modified adenovirus vectorsmediate efficient gene transfer into undifferentiated and adipogenic-differentiated humanmesenchymal stem cells. Biochem Biophys Res Commun. 2005; 332:1101–1106. [PubMed:15922299]

9. Derugin N, Ferriero DM, Vexler ZS. Neonatal reversible focal cerebral ischemia: a new model.Neurosci Res. 1998; 32:349–353. [PubMed: 9950062]

10. van Velthoven CT, Kavelaars A, van Bel F, Heijnen CJ. Nasal administration of stem cells: apromising novel route to treat neonatal ischemic brain damage. Pediatr Res. 2010; 68:419–422.[PubMed: 20639794]

11. Nijboer CH, van der Kooij MA, van Bel F, Ohl F, Heijnen CJ, Kavelaars A. Inhibition of the JNK/AP-1 pathway reduces neuronal death and improves behavioral outcome after neonatal hypoxic-ischemic brain injury. Brain Behav Immun. 2010; 24:812–821. [PubMed: 19766183]

12. Faustino JV, Wang X, Johnson CE, Klibanov A, Derugin N, Wendland MF, et al. Microglial cellscontribute to endogenous brain defenses after acute neonatal focal stroke. J Neurosci. 2011;31:12992–13001. [PubMed: 21900578]

13. Zhang ZG, Chopp M. Neurorestorative therapies for stroke: underlying mechanisms andtranslation to the clinic. Lancet Neurol. 2009; 8:491–500. [PubMed: 19375666]

14. Torrente Y, Polli E. Mesenchymal stem cell transplantation for neurodegenerative diseases. CellTransplant. 2008; 17:1103–1113. [PubMed: 19181205]

15. Kim ES, Ahn SY, Im GH, Sung DK, Park YR, Choi SH, et al. Human umbilical cord blood-derived mesenchymal stem cell transplantation attenuates severe brain injury by permanent middlecerebral artery occlusion in newborn rats. Pediatr Res. 2012; 72:277–284. [PubMed: 22669296]

16. Ohtaki H, Ylostalo JH, Foraker JE, Robinson AP, Reger RL, Shioda S, et al. Stem/progenitor cellsfrom bone marrow decrease neuronal death in global ischemia by modulation of inflammatory/immune responses. Proc Natl Acad Sci USA. 2008; 105:14638–14643. [PubMed: 18794523]

17. Geddes R, Vannucci RC, Vannucci SJ. Delayed cerebral atrophy following moderate hypoxia-ischemia in the immature rat. Dev Neurosci. 2001; 23:180–185. [PubMed: 11598317]

18. Levison SW, Goldman JE. Both oligodendrocytes and astrocytes develop from progenitors in thesubventricular zone of postnatal rat forebrain. Neuron. 1993; 10:201–212. [PubMed: 8439409]

van Velthoven et al. Page 8

Stroke. Author manuscript; available in PMC 2014 May 01.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 9: free article

19. Yang Z, Covey MV, Bitel CL, Ni L, Jonakait GM, Levison SW. Sustained neocorticalneurogenesis after neonatal hypoxic/ischemic injury. Ann Neurol. 2007; 61:199–208. [PubMed:17286251]

20. Donega V, van Velthoven CT, Nijboer CH, van Bel F, Kas MJ, Kavelaars A, et al. Intranasalmesenchymal stem cell treatment for neonatal brain damage: long-term cognitive andsensorimotor improvement. PLoS One. 2013; 8:e51253. [PubMed: 23300948]

21. Gonzalez FF, Abel R, Almli CR, Mu D, Wendland M, Ferriero DM. Erythropoietin sustainscognitive function and brain volume after neonatal stroke. Dev Neurosci. 2009; 31:403–411.[PubMed: 19672069]

22. Jiang Y, Wei N, Lu T, Zhu J, Xu G, Liu X. Intranasal brain-derived neurotrophic factor protectsbrain from ischemic insult via modulating local inflammation in rats. Neuroscience. 2011;172:398–405. [PubMed: 21034794]

23. Schabitz WR, Steigleder T, Cooper-Kuhn CM, Schwab S, Sommer C, Schneider A, et al.Intravenous brain-derived neurotrophic factor enhances poststroke sensorimotor recovery andstimulates neurogenesis. Stroke. 2007; 38:2165–2172. [PubMed: 17510456]

24. Ploughman M, Windle V, MacLellan CL, White N, Dore JJ, Corbett D. Brain-derived neurotrophicfactor contributes to recovery of skilled reaching after focal ischemia in rats. Stroke. 2009;40:1490–1495. [PubMed: 19164786]

25. Park H, Poo MM. Neurotrophin regulation of neural circuit development and function. Nat RevNeurosci. 2012; 14:7–23. [PubMed: 23254191]

26. Borlongan CV, Weiss MD. Baby STEPS: a giant leap for cell therapy in neonatal brain injury.Pediatr Res. 2011; 70:3–9. [PubMed: 21659957]

27. Ming GL, Song H. Adult neurogenesis in the mammalian brain: significant answers and significantquestions. Neuron. 2011; 70:687–702. [PubMed: 21609825]

van Velthoven et al. Page 9

Stroke. Author manuscript; available in PMC 2014 May 01.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 10: free article

Figure 1. Effect of MSC transplantation on lesion sizeQuantification of damage using Nissl staining (A), MAP2-positive area loss (B), MBP-positive area loss (C) and representative sections. Data represent mean±SEM. Sham controlsn=5, VEH n=8, MSC n=9, MSC-BDNF n=8. *p<0.05 vs. VEH, **p<0.01 vs. VEH.

van Velthoven et al. Page 10

Stroke. Author manuscript; available in PMC 2014 May 01.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 11: free article

Figure 2. Effect of MSC transplantation on functional outcomeRats underwent MCAO at p10 and were treated with vehicle or MSC at 3 days post-MCAo.Lateralizing motor deficits were measured in the cylinder rearing test at 14, 21 and 28 dayspost-MCAo (A) and using the adhesive removal test at 28 days post-MCAo (B). Datarepresent mean±SEM. Sham-controls n=5, VEH n=8, MSC n=9, MSC-BDNF n=8. *p<0.05vs. VEH, #p<0.05 MSC-BDNF vs. MSC.

van Velthoven et al. Page 11

Stroke. Author manuscript; available in PMC 2014 May 01.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 12: free article

Figure 3. Effect of MSC transplantation on cell proliferation in ipsilateral striatum andsubventricular zoneThe number of EdU-positive cells (A) and BrdU-positive cells (B) was measured in theipsilateral striatum. Cell proliferation in the SVZ at 28 days post-MCAO was measuredusing Ki67 (C). Panel D shows representative photomicrographs of EdU-, BrdU- and Ki67-labelled cells. Scale bar represents 50µm. Data represent mean±SEM. Sham controls n=5,VEH n=8, MSC n=9, MSC-BDNF n=8. *p<0.05 vs. VEH, **p<0.01 vs. VEH.

van Velthoven et al. Page 12

Stroke. Author manuscript; available in PMC 2014 May 01.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript