You are on page 1of 13

Brain, Behavior, and Immunity 70 (2018) 118–130

Contents lists available at ScienceDirect

Brain, Behavior, and Immunity


journal homepage: www.elsevier.com/locate/ybrbi

Full-length Article

Interaction between hypothermia and delayed mesenchymal stem cell


therapy in neonatal hypoxic-ischemic brain injury
Josephine Herz a,⇑, Christian Köster a,b, Barbara S. Reinboth a, Mark Dzietko a, Wiebke Hansen c,
Hemmen Sabir a, Cindy van Velthoven d, Ivo Bendix a,b,1, Ursula Felderhoff-Müser a,⇑,1
a
Department of Pediatrics 1/Neonatology University Hospital Essen, University of Duisburg-Essen, Essen, Germany
b
Experimental Perinatal Neuroscience, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
c
Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
d
Department of Neurology and Neurological Sciences, Stanford University, Stanford, USA

a r t i c l e i n f o a b s t r a c t

Article history: Acute hypothermia treatment (HT) is the only clinically established intervention following neonatal
Received 18 September 2017 hypoxic-ischemic brain injury. However, almost half of all cooled infants still die or suffer from long-
Received in revised form 2 February 2018 lasting neurological impairments. Regenerative therapies, such as mesenchymal stem cells (MSC) appear
Accepted 13 February 2018
promising as adjuvant therapy. In the present study, we hypothesized that HT combined with delayed
Available online 14 February 2018
MSC therapy results in augmented protection, improving long-term neurological outcome. Postnatal
day 9 (P9) C57BL/6 mice were exposed to hypoxia–ischemia followed by 4 h HT. Murine bone
Keywords:
marrow-derived MSC (1  106 cells/animal) were administered intranasally at P12. Cytokine and growth
Neonatal hypoxia–ischemia
Perinatal asphyxia
factor levels were assessed by ELISA and LuminexÒ multiplex assay 24 h following MSC delivery. One
Therapeutic hypothermia week after HI, tissue injury and neuroinflammatory responses were determined by immunohistochem-
Mesenchymal stem cells istry and western blot. Long-term motor-cognitive outcome was assessed 5 weeks post injury. MSC
Long-term functional outcome responses to the brains’ environment were evaluated by gene expression analysis in MSC, co-cultured
Neuroinflammation with brain homogenates isolated at P12. Both, MSC and HT improved motor deficits, while cognitive func-
tion could only be restored by MSC. Compared to each single therapy, combined treatment led to
increased long-lasting motor-cognitive deficits and exacerbated brain injury, accompanied by enhanced
endothelial activation and peripheral immune cell infiltration. MSC co-cultured with brain extracts of HT-
treated animals revealed increased pro-inflammatory cytokine and decreased growth factor expression.
In vivo protein analysis showed higher pro-inflammatory cytokine levels after combined treatment com-
pared to single therapy. Furthermore, HI-induced increase in growth factors was normalized to control
levels by HT and MSC single therapy, while the combination induced a further decline below control
levels. Our results suggest that alteration of the brains’ microenvironment by acute HT modulates MSC
function resulting in a pro-inflammatory environment combined with alteration of the homeostatic
growth factor milieu in the neonatal hypoxic-ischemic brain. This study delineates potential unexpected
side effects of cell-based therapies as add-on therapy for acute hypothermia treatment.
Ó 2018 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).

1. Introduction ing in cerebral palsy, epilepsy, visual impairment and motor-


cognitive deficits in later life (Eunson, 2015).
Perinatal asphyxia, resulting in hypoxic-ischemic encephalopa- To date, therapeutic hypothermia (HT) is the only recom-
thy (HIE), is one of the worldwide leading causes of death and mended therapeutic intervention following perinatal asphyxia
disability in children. In high-income countries 1 to 6 per 1000 (ILCOR guidelines 2015). Neuroprotective mechanisms involved
newborns suffer from HIE during the perinatal period often result- are inhibition of acute metabolic disturbance, inhibition of apop-
totic cell death and suppression of inflammation (Wassink et al.,
2014). Nevertheless, 40–50% of cooled infants still suffer from
⇑ Corresponding authors at: Department of Pediatrics 1, University Hospital
major neurological problems and the therapeutic window of
Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Essen, Germany.
opportunity (6 h) is challenging (Azzopardi et al., 2014). Experi-
E-mail addresses: josephine.herz@uk-essen.de (J. Herz), ursula.felderhoff@uk-
essen.de (U. Felderhoff-Müser). mentally, we and others demonstrated short-term neuroprotection
1
Equally contributing senior authors. by HT following HIE-induced brain injury, whereas long-term

https://doi.org/10.1016/j.bbi.2018.02.006
0889-1591/Ó 2018 The Authors. Published by Elsevier Inc.
This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
J. Herz et al. / Brain, Behavior, and Immunity 70 (2018) 118–130 119

neurodevelopment is only partially restored, closely resembling conjugated anti-CD45 (BD Biosciences, Germany, clone: 30-F11),
the clinical situation (Azzopardi et al., 2014; Burnsed et al., 2015; anti-TER119 (eBiosciences, Germany, clone: TER-119), anti-
Pappas et al., 2015; Reinboth et al., 2016). CD11b (BD Biosciences, clone: M1/70), anti-MHC II (eBiosciences,
Considering limited long-term neurodevelopmental improve- clone: M5/114.15.2), anti-Sca-1 (eBiosciences, clone: D7), anti-
ment following HT, combined treatment with regenerative cell- CD29 (eBiosciences, clone: eBioHMb-1(HMB1-1)), anti- CD44
based therapies attenuating sub-acute injury processes and pro- (eBiosciences, clone: IM7) and anti-CD49e (eBiosciences, clone:
moting repair appears promising. To increase endogenous repair, eBioHMa5-1 (HMa5-1)) antibodies for 20 min at 4 °C. Isotype-
bone marrow-derived mesenchymal stem cells (MSC) have gained matched control monoclonal antibodies were used to set appropri-
major interest in the past. Pioneer work in a murine model of ate gates for positive staining considering the level of background
neonatal HIE by van Velthoven et al. revealed that MSC provide staining (Supplementary Fig. S1A). Flow cytometric analyses were
long-term neuroprotection through promotion of endogenous cell performed on a BD LSRII equipped with FACS Diva Software (both
proliferation, inhibition of microglia activation and preservation BD Biosciences) which was used for acquisition and data analysis.
of white and grey matter (van Velthoven et al., 2010a). Recent evi- Osteogenic and adipogenic differentiation potential (Supple-
dence also suggests that intranasal MSC administration is an effi- mentary Fig. S1B) of cultured MSC was evaluated by seeding 5 x
cient and less invasive route to treat neonatal brain injury (van 104 MSC per well in a 24-well culture plate (duplicates) with
Velthoven et al., 2010a). Furthermore, in rodents, the therapeutic MSC growth medium (DMEM low glucose GlutaMAXTm,
time window of intranasal MSC therapy has been reported to be Gibco)/20% fetal calf serum (FCS, Biochrom). After 48 – 72 h
expandable for up to 10 days post HIE (Donega et al., 2013). (100% confluence) one of each duplicate samples was exposed to
Delayed treatment options will provide time for logistics (e.g. MSC osteogenic or adipogenic differentiation medium (PromoCell,
transferring patients from peripheral hospitals for treatment, con- Germany). MSC growth medium was used for the remaining wells
firmed diagnosis of HIE by clinical course, neurophysiology and as negative control. Cells were incubated for 18–21 days with
imaging). Furthermore, because of the short therapeutic window, media exchange every third day. Osteocyte differentiation was
HT is supposed to target early pathophysiological processes. In analyzed via Alizarin Red (2%) staining for 45 min. Differentiation
contrast, MSC have been shown to act on secondary injury pro- into adipocytes was confirmed by Oil-Red (0.3%) staining for 15
cesses and promote endogenous repair which is initiated in the min (Supplementary Fig. S1B).
post-acute phase (Ferriero, 2004; Gunn et al., 2017; Thornton Chondrogenic differentiation potential was determined by
et al., 2012). Therefore, acute HT with delayed MSC therapy would seeding 4x105 cells in 15 ml Falcon tubes in duplicate to enable
be an attractive therapy design. formation of spheroids. After 24–48 h one sample of duplicates
Even though small clinical trials (e.g. NCT02612155, was exposed to chondrogenic differentiation medium (PromoCell)
NCT02881979), investigating the effect of stem cell therapy in while MSC growth medium was used as negative control in the
cooled asphyxiated newborns, are already underway, pre-clinical remaining tube. Medium was exchanged every 3 days. After incu-
studies have rarely addressed the benefit of the combined treat- bation for 18–21 days cell pellets were carefully removed and fro-
ment. Park et al. delivered human umbilical cord blood MSC intra- zen on dry ice. Cell pellets were cut into 10 mm cryostat sections
ventricularly prior to HT, resulting in long-term protection from and differentiation into chondrocytes was evaluated by staining
HIE-induced brain tissue loss (Park et al., 2015). However, an acute with Alcian Blue (0.25%) for 5 h (Supplementary Fig. S1B).
intracerebral cell administration is unlikely to be feasible in the
clinical setup following perinatal asphyxia. Experimental work
investigating HT in combination with a less invasive and delayed 2.3. Animal care and group allocation
stem cell therapy was lacking.
The present study investigated the hypothesis that immediate Experiments were performed in accordance to the Animal
HT combined with delayed intranasal MSC therapy promotes Research: Reporting of In Vivo Experiments (ARRIVE) guidelines
endogenous repair mechanisms and inhibits secondary injury pro- with government approval by the State Agency for Nature, Envi-
cesses overcoming current limitations of HT for the treatment of ronment and Consumer Protection North Rhine-Westphalia.
neonatal HIE. C57BL/6J mice were bred in house and kept under a 12 h light/dark
cycle with food and water ad libitum. Animals subjected to behav-
ioral testing were weaned at postnatal day 21 (P21) and housed in
2. Materials and methods
groups of 4–5 animals per cage. Bodyweight was recorded at P10,
P11, P12, P16 and once a week after weaning.
2.1. Isolation and culture of murine bone marrow mesenchymal stem
A total of 305 P9 pups (n = 151 female and n = 154 male)
cells
derived from 41 litters were enrolled. Forty-seven mice underwent
sham surgery. Out of the 258 animals exposed to hypoxia-ischemia
Bone marrow MSC were cultured according to previously pub-
18 animals (7%) died immediately after or during hypoxia. Temper-
lished protocols (van Velthoven et al., 2010a). Briefly, bone marrow
ature during intervention was monitored in sentinel pups (n = 42
from femurs and tibias of 5–7 week old C57BL/6J mice (Harlan,
in total, Supplementary Fig. S2A), being excluded from data analy-
Germany) was flushed with Dulbeccos modified eagle medium
sis to minimize confounding effects by stressful temperature mea-
low glucose GlutaMAXTm (DMEM, Gibco, Germany) /20% fetal calf
surements. Remaining animals were randomly assigned to 4
serum (FCS, Biochrom, Germany). Cells were sub-cultured four to
treatment groups by an independent scientist not involved in data
six times prior to administration. For in vivo tracking analysis,
acquisition: (1) hypoxia ischemia (HI)/normothermia (NT, n = 18
GFP-labelled MSC (Cyagen Biosciences Inc, USA) were thawed
(5 weeks survival), n = 16 (1 week survival), n = 8 (4 days survival),
and sub-cultured twice before intranasal delivery.
n = 8 (3 days survival)), (2) HI/hypothermia (HT, n = 20 (5 weeks
survival), n = 14 (1 week survival), n = 8 (4 days survival), n = 8
2.2. Phenotypic characterization of cultured murine bone marrow- (3 days survival)), (3) HI/NT/MSC (MSC, n = 17 (5 weeks survival),
derived mesenchymal stem cells n = 16 (1 week survival), n = 16 (4 days survival)) (4) HI/HT/MSC
(HT + MSC, n = 18 (5 weeks survival), n = 15 (1 week survival),
Cultured MSC were stained with fluorescein isothiocyanate, n = 16 (4 days survival)). In total 1 NT and 1 HT + MSC died within
phycoerythrine, allophycocyanin, Alexa-Flour 780 or eF450- the first week after HI; 2 HT and 2 HT + MSC animals died between
120 J. Herz et al. / Brain, Behavior, and Immunity 70 (2018) 118–130

1 and 5 weeks post injury induction. Experimenters were blinded maze followed by one day open field and one day novel object
to all interventions and data analysis. recognition test. Data were recorded using an automatic tracking
system (Video-Mot2, TSE Systems, Germany) and exported for sta-
2.4. Neonatal hypoxia-ischemia tistical analysis. All experimental procedures were carried out in
the dark phase in a dimly lit (red light) and a low noise environ-
Hypoxic-ischemic (HI) brain injury was induced as previously ment (behavioral unit).
described (Reinboth et al., 2016). Briefly, the right common carotid
artery was occluded through cauterization (high temperature cau- 2.7. Tissue preparation, histology and immunohistochemistry
ter, 1200 °C, Bovie, USA) under isoflurane anesthesia (1.5–4 vol%,
total duration of surgery: 5–7 min) followed by one hour hypoxia One week after HI, mice were deeply anesthetized with chloral-
(10% O2) in an air-tight oxygen chamber (OxyCycler, Biospherix, hydrate (200 mg/kg body weight) and transcardially perfused with
USA) after one hour recovery with their dams. According to our ice-cold phosphate buffered saline (PBS). Brains were removed and
previous study where we identified a physiological body core (i.e. snap frozen on dry ice. Tissue injury was assessed and scored on
nesting) temperature of 35 °C for P9 mice (Reinboth et al., 2016) cresyl violet stained 20 mm cryostat sections as previously
animals were placed on a warming mat (Harvard Apparatus, described (Reinboth et al., 2016; Sheldon et al., 1998) with minor
USA) to maintain nesting temperature during hypoxia. Sham- modifications. Briefly, 9 regions were scored: the anterior, middle
operated animals received anesthesia and neck incision only. and posterior cortex, CA1, CA2, CA3 and dentate gyrus of the hip-
pocampus, the striatum and the thalamus. Each region was given
2.5. Therapeutic hypothermia and MSC-treatment a rating from 0 to 3 (0- no detectable cell loss, 1- small focal areas
of neuronal cell loss, 2- columnar damage in the cortex or moder-
According to our previous study, HT was applied immediately ate to severe cell loss in the other regions, 3- cystic infarction and
following HI for 4 h with a target temperature of Trectal 32 °C gliosis). The sum score from different regions was calculated for
(Reinboth et al., 2016). Mice were placed on a custom-made plate each animal resulting in a total maximum score of 27.
with temperature control by water circulation. Controls (NT) were Cellular injury and neuroinflammatory responses were assessed
placed on a warming mat to maintain physiological body core tem- on cryostat sections from the striatal (+0.2 to +0.3 mm from
perature (Trectal 35 °C) conforming to our previous work (Reinboth bregma) and the hippocampal (1.9 to 2.0 mm from bregma)
et al., 2016). Three days after HI (Fig. 1A), a total of 1x106 MSC or level via immunohistochemical detection of neurons (NeuN), oligo-
vehicle (0.9% sodium chloride) were administered intranasally as dendrocytes (Olig2), leukocytes (CD45) and vessels (CD31) accord-
previously described (Donega et al., 2013; van Velthoven et al., ing to previously published protocols (Herz et al., 2015; Reinboth
2010a). This treatment regime aimed to extend the therapeutic et al., 2016). Briefly, brain tissue sections (20 mm) were thawed
time window and was selected based on a previous study reporting and dried at 37 °C followed by fixation in ice-cold 4%
pronounced neuroprotection after delivery of 1x106 cells at 3 days paraformaldehyde (for NeuN, Olig2) or aceton/methanol (1:1 v/v
post HI (Donega et al., 2013). %, for CD45, CD31) for 10 min at 4 °C. Unspecific antibody binding
was blocked by incubation with 1% bovine serum albumin (BSA,
2.6. Long-term functional outcome Sigma-Aldrich), 0.3% cold fish skin gelatin (Sigma Aldrich), 0.05%
Tween 20 (Sigma Aldrich) in Tris-buffered saline (TBS, for NeuN,
Behavioral assessment was performed according to our previ- Olig2) or 5% donkey, 5% normal goat serum, 2% BSA, 0.2% Tween
ously described protocols (Reinboth et al., 2016). Shortly, explo- 20 in PBS (for CD45, CD31) for 1 h at room temperature. Sections
ration and impulsive behavior was assessed in the elevated plus were incubated with the following primary antibodies: rabbit
maze (Lister, 1987; Walf and Frye, 2007). Mice were placed on anti-mouse Olig2, (1:100, Millipore, Germany); rabbit anti-mouse
the central platform and behavior was recorded for 5 min. The NeuN, (1:500, Millipore), rat anti-mouse CD45 (1:20, BD Pharmin-
time spent in the open arms and the time of head-dipping in open gen, Germany), rat-anti-mouse CD31 (1:500, BD Pharmingen) in
and center regions was measured. The open field test was used to blocking solution at 4 °C overnight. Antibody binding was
evaluate spontaneous motor- and rearing activity and to assess visualized by incubation with appropriate secondary antibodies
anxiety/exploration-related behavior (Milner and Crabbe, 2008). (anti-rabbit or anti-rat Alexa Fluor 488, both: 1:500, Invitrogen,
Animals were placed into the center of an open field arena and Germany) for 2 h at room temperature. Nuclei were counterstained
movements were recorded by the tracking system for 5 min. with 40 ,6-Diamidin-2-phenylindol (Dapi, 100 ng/ml; Molecular
Besides measurement of mean velocities, anxiety/exploration- Probes, USA).
related behavior was determined as the percentage of time the
animal stayed in the central area of the box in relation to the total 2.8. Quantification of neuronal, oligodendrocyte and vessel density;
time spent in the arena. Rearing activity was calculated as the vessel diameters and peripheral leukocyte infiltration
time the animals reared in the border region relative to the total
time the mice spent in this region. The novel object recognition Defined non-overlapping regions of interest (ROI, each 1,89,000
task is a non-spatial, non-aversive memory test which relies on mm2 (CD45, CD31) and 45,900 mm2 (NeuN, Olig2)) were visualized
the observation, that animals preferentially explore novel objects by fluorescence microscopy (20 and 40 objective; Axioplan;
over those that are familiar (Bevins and Besheer, 2006). In the ini- Zeiss, Germany) connected to a CCD camera (Axiocam ICc1, Zeiss).
tial familiarization trial, animals were placed in the open field For representative images, low and high magnification images
arena, with 2 identical objects in two facing corners. The animals were acquired with confocal microscopy (A1plus, Eclipse Ti, with
were returned to their cages for an inter-trial interval of 30 min. NIS Elements AR software, Nikon, Germany). At the level of the
In the following retention/test trial, animals were exposed to striatum 6 defined non-overlapping ROI were analyzed in each
one familiar object and one novel object replacing the second hemisphere (3 ROI in the cortex and 3 ROI in the striatum). At
familiar object in the arena. Novel object activity was recorded the level of the hippocampus 10 ROI (3 ROI in the cortex, 4 ROI
for 5 min. in the hippocampus and 3 ROI in the thalamus) were evaluated
From P21 onwards, animals were transferred to an inversed in each hemisphere. Oligodendrocytes and vessels were counted
12 h light/dark cycle and behavioral testing was initiated at P44 manually on acquired images. Since single cell counting could
(Reinboth et al., 2016). It started with one day of elevated plus not be applied in densely packed regions of neurons or strong
J. Herz et al. / Brain, Behavior, and Immunity 70 (2018) 118–130 121

Fig. 1. Improved long-term functional outcome by acute hypothermia and delayed intranasal MSC treatment is reduced after combination of both therapies. (A) Experimental
paradigm and readouts. (B–D) Behavioral testing was initiated on P44 in young adult animals that were either sham-operated or exposed to HI followed by 4 h NT or HT on
P9. MSC were administered intranasally at P12. (B) In the Elevated plus maze the time spent in open arms and time of head-dipping in open and center regions was measured.
(C) Rearing activity was quantified in the Open field maze by calculation of the time animals reared in the border region relative to total time mice spent in this region.
(D) Cognitive function was assessed by Novel object recognition task where animals were exposed to one familiar object and one novel object after a training trial with
two identical objects. Percentage of time mice spent with the new object of total object time was analyzed. n = 16–18/group, *p < 0.05, **p < 0.01, ***p < 0.001,
NT = normothermia, HT = hypothermia, MSC = mesenchymal stem cells.

leukocyte infiltration (e.g. hippocampus), images were converted were measured with Image J for 12–15 randomly selected vessels
into binary images followed by measurement of NeuN and CD45 per animal derived from three non-overlapping hippocampal ROI
positive areas using Image J (NIH, USA). Ipsilateral vessel diameters (CA1, CA2, DG covering a total area of 0.567 mm2).
122 J. Herz et al. / Brain, Behavior, and Immunity 70 (2018) 118–130

2.9. Western blot million MSC/well were pre-cultured in 6-well plates for 24 h in
DMEM/20% FCS followed by culture in DMEM knock out medium
For western blot analysis sections of 200 mm thickness of the supplemented with isolated brain extract (1 mg protein/ml med-
ipsilateral hemisphere within the range of 0.3 to 0 mm from ium). After 48 h total RNA was isolated with the RNeasy Micro
bregma (striatal level) and 2.0 to 2.3 mm from bregma (hip- Kit (Qiagen Germany) according to the manufactures recommen-
pocampal level) were dissected and homogenized in ice-cold lysis dations. First strand complementary DNA was synthesized using
buffer (RIPA, Sigma-Aldrich) containing protease and phosphatase 1 mg of total RNA and TaqMan reverse transcription reagents
inhibitors (cOmplete, Roche) and 100 mM PMSF (Sigma-Aldrich). (Applied Biosystems/Thermo Fisher Scientific USA). PCR amplifica-
The supernatant was collected and processed as previously tion was performed in 96 well optical reaction plates for 40 cycles
described (Reinboth et al., 2016). Briefly, after determination of with each cycle at 94 °C for 15 s and 60 °C for 1 min using the Ste-
protein concentration using the Pierce bicinchoninic acid assay pOnePlus Real Time PCR system (Applied Biosystems/Thermo
(Thermo Scientific, USA), protein lysates were separated on gradi- Fisher Scientific). PCR products of tumor necrosis factor alpha
ent sodiumdodecylsulphate polyacrylamide gels (Mini-PROTEAN (tnf-alpha, Mm00443258_m1), interleukin (IL)-1beta (il-1beta,
TGX Precast Gels, Any kDa, BioRad, Germany) and transferred to Mm00434228_m1)), il-6 (Mm00446190_m1); il-12
nitrocellulose membranes (0.2 mm, Amersham, USA) at 4 °C over- (Mm00434169_m1), il-10 (Mm01288386_m1), brain derived neu-
night. Equal loading of 20 mg/lane and transfer of proteins was con- rotrophic factor (bdnf, Mm01334043_m1), epidermal growth factor
firmed by staining of membranes with Ponceau S solution (Sigma- (egf, Mm00438696_m1), insulin like growth factor (igf,
Aldrich). Nonspecific binding was blocked by incubation in 5% non- Mm00439560_m1), vascular endothelial growth factor (vegf,
fat milk powder, 0.1% Tween in TBS (TBST) followed by incubation Mm00437306_m1) and beta-2-micorglobulin (Mm00437762_m1)
with the primary antibodies, i.e. rabbit anti-myelin basic protein were quantified using assay on demand primers and fluorogenic
(MBP, 1:10.000, Covance, USA), mouse anti-microtubuli associated reporter oligonucleotide probes (Applied Biosystems/Thermo
protein-2 (MAP-2, 1:1000, Sigma-Aldrich), rabbit anti-ionized cal- Fisher Scientific). CT values were normalized for the housekeeping
cium binding adaptor molecule-1 (Iba-1, 1:1000, Wako, Japan), gene beta-2-microglobulin [DCT = CT (target gene)-CT (beta-2-
goat anti-vascular cell adhesion molecule-1 (VCAM-1, 1:10.000, microglobulin)] and related to the mean of control samples (MSC
R&D Systems, USA) and rabbit anti-glutaraldehyde-3-phosphate co-cultured with sham brain extracts) using the DDCT formula
dehydrogenase (GAPDH, 1:1000, Santa Cruz, USA) in blocking solu- [DDCT = DCT (sample) -DCT (control)]. Fold change values were
tion at 4 °C overnight. Membranes were incubated with appropri- calculated.
ate peroxidase-conjugated secondary antibodies (all 1:2000 except
of anti-goat horseradish peroxidase (1:10.000), all Dako, Denmark) 2.12. Assessment of cytokines and growth factors
in blocking solution at room temperature for 1 h followed by
Chemiluminescent detection with the enhanced chemilumines- Abundance of cytokines and growth factors was assessed in
cence prime western blotting detection reagent (Amersham, GE brain lysates, plasma samples and supernatants of MSC co-
Healthcare Life Science, USA). For visualization and densitometric cultures. For analysis of brain tissues, mice were perfused, hemi-
analysis the ChemiDocXRS+ imaging system and ImageLab soft- spheres dissected (excluding cerebellum), snap frozen on dry ice
ware (Bio-Rad, Germany) were used. and stored at 80 °C until further processing. Tissues were homog-
enized in ice-cold Cell Lysis Buffer 2 (R&D Systems, Germany).
2.10. MSC tracking analysis Blood was collected from the right atrium prior to perfusion and
collected in ethylenediaminetetraacetate (EDTA) coated tubes.
According to Donega et al. MSC homing was analyzed at 14–16 Samples were centrifuged at 2000g for 20 min and the super-
h following intranasal MSC delivery 3 days after HI (Donega et al., natant was frozen at 80 °C until further analysis. Cell culture
2014). The amount of GFP-labelled MSC in brains of NT and HT- supernatants, brain lysates and plasma samples were assayed for
treated animals was quantified by flow cytometry. Isolation of sin- the presence of TNFalpha, IL-1beta, IL-6, IL-12, IL-10 and VEGF per-
gle cell suspension for flow cytometry analysis was performed as forming a LuminexÒ multiplex screening assay (R&D Systems)
previously described (Herz et al., 2014). Hemispheres were dis- according to the manufacturers’ instructions. Samples were ana-
sected, cerebellum removed followed by homogenization through lyzed by Luminex 200 technology using Luminex IS software
a 70 lm cell strainer (BD Biosciences, Germany) during continuous (Luminex Corporation, USA). In brain lysates concentrations of IL-
rinsing with 15 ml of cold HEPES-buffered RPMI1640. Two hemi- 12, in plasma samples concentrations of IL-1beta, IL-6, IL-12 and
spheres were pooled per sample to avoid potential cell loss in culture supernatants concentrations of IL-1beta, IL-12, IL-6 and
through tissue processing. Samples were centrifuged at 400g VEGF were out of the detection range of the multiplex assay. Pro-
for 10 min at 18 °C. The supernatants were discarded and the pel- tein abundance for BDNF, EGF and IGF was determined by ELISA
lets were re-suspended in 37% Percoll followed by centrifugation at (R&D Systems) according to manufacturers’ instructions.
2800g for 20 min. Myelin was removed and the remaining cell
pellet was washed twice in PBS. Total cell counts were determined 2.13. Statistical analysis
using BD TrueCount beads (BD Biosciences). Viable cells were
quantified by adding 1 mg/ml propidium iodide immediately prior All results are presented as scatter blots with bars presenting
to measurement. Data acquisition and analysis were performed mean values unless stated otherwise. Sample size was determined
on a BD FACS LSRII equipped with FACS Diva software (BD a priori using G⁄Power (version 3.1). An a-level of 0.05 and a
Biosciences). power of 0.8 were required. Based on our previous report
(Reinboth et al., 2016), a Cohen’s d ES of 0.5 and a mortality of
2.11. MSC co-culture with brain extracts and gene expression analysis 15% was assumed yielding a final sample size of at least 14 animals
per group for sub-acute brain injury and long-term functional out-
MSC were cultured with brain extracts as previously described come. For statistical analysis, the GraphPad Prism 6.0 software
(van Velthoven et al., 2010b). Tissues of ipsilateral hemispheres package (GraphPad Software, USA) was used. After checking for
(+0.5 to 2.3 mm from bregma) of mice from all treatment groups normal distribution either ordinal 1-way ANOVA with post hoc
were dissected on ice followed by homogenization in knock-out Bonferroni multiple comparisons test or Kruskal-Wallis with
DMEM (Gibco). Supernatants were stored at -80 °C until use. One Dunn’s multiple comparisons test were applied. For comparison
J. Herz et al. / Brain, Behavior, and Immunity 70 (2018) 118–130 123

of two groups either Students’ t-test or Mann Whitney U test was treatment leading to a significantly reduced novel object activity
used. A p value of <0.05 was considered significant. compared to sham controls (Fig. 1D).

3.2. Combining acute hypothermia with delayed MSC treatment


3. Results diminishes early neuroprotection

3.1. Improved long-term neurological function by acute hypothermia To assess whether the observed detrimental interaction was ini-
and delayed intranasal MSC treatment is decreased following tiated early after combining both treatments, we analyzed
combined treatment histopathological, cellular and structural grey and white matter
injury at one week post HI.
Using our established HI/HT protocol for neonatal C57BL/6 mice Total and local hippocampal injury was significantly reduced by
we confirmed that the previously determined nesting temperature both, HT and MSC (Fig. 2). After combining both treatments, pro-
of 35 °C and the target temperature difference of 3 °C (Reinboth tective effects were decreased resulting in significantly increased
et al., 2016) was maintained during experimental modelling injury scores of HT + MSC compared to single MSC therapy for total
(Supplementary Fig. S2A). HI animals revealed significantly and local injury within the hippocampus (Fig. 2). In cortex and
reduced body weight gain compared to sham-operated animals striatum, HT- and MSC-induced significant protection was
early after HI independent of treatment (Supplementary reversed after combined treatment (Supplementary Fig. S4A, B).
Fig. S2B). Confounding effects on neurobehavioral assessment by In the thalamus, all administered therapies revealed similar pro-
different weight gain could be excluded because no significant tection (Supplementary Fig. S4C).
differences were observed across all experimental groups 5 weeks For further insight into target structures with respect to sub-
after HI (Supplementary Fig. S2B). acute grey and white matter injury, western blot analyses for
Exploratory and impulsive behavior, analyzed by elevated plus MAP-2 and MBP expression at the level of the striatum and the hip-
maze, demonstrated that both single therapies (HT and MSC treat- pocampus were performed. NT animals demonstrated a marked
ment) reduced HI-induced alterations with no significant differ- loss of both axonal and myelin structures in ipsilateral hemi-
ences compared to sham-operated control mice (Fig. 1B). spheres (Fig. 3). Whereas HT significantly improved expression of
However, combined treatment reversed protective effects by HT MAP-2 expression, MSC treatment did not have an effect on axonal
and MSC leading to significantly increased time intervals mice degeneration (Fig. 3). In contrast, myelination assessed by quantifi-
spent in the open arms compared to sham mice (Fig. 1B). A compa- cation of MBP expression was significantly improved by MSC treat-
rable regulation was observed for the time of head-dipping in the ment but not by HT (Fig. 3). Interestingly, combination of HT and
open and center regions of the maze (Fig. 1B). General motor activ- MSC resulted in a significant improvement of MAP-2 expression
ity assessed in the open field maze was not significantly modulated at the hippocampal level, whereas enhanced myelination by MSC
by either treatment (Supplementary Fig. S3A). Similar to observa- was abolished in animals treated with HT before MSC administra-
tions made in the elevated plus maze HT + MSC animals spent tion (Fig. 3).
longer times in the center region of the open field maze (Supple- To analyze whether differential effects on structural damage
mentary Fig. 3B). HT significantly improved vertical activity, as a might be attributed to cellular loss or differentiation deficits, we
measure of motor function, which was diminished after combina- quantified neuronal and oligodendrocyte density via immunohis-
tion with MSC therapy (Fig. 1C). Cognitive function, evaluated by tochemistry for NeuN and Olig2. The most pronounced reduction
novel object recognition was reduced in NT mice and was not of neuronal density was observed in the hippocampus and stria-
recovered by HT. However, MSC significantly improved long-term tum, which was ameliorated by single HT and MSC treatment
cognitive function (Fig. 1D), being reversed when HT preceded (Fig. 4A, B). However, after combining both therapies the single

Fig. 2. Combining acute hypothermia with delayed MSC treatment diminishes protective effects on sub-acute histological brain injury. Histological brain injury was
determined on cresyl violet stained 20 mm cryostat sections of P16 mice that were exposed to HI followed by 4 h NT or HT on P9. MSC were administered intranasally at P12.
(A) Representative images of injured hemispheres for each experimental group are shown (scale bar: 1 mm). (B) Pictures display higher magnification images (scale bar:
100 mm) of squares depicted in (A) revealing severe cell loss throughout the pyramidal cell layer of the hippocampus in NT and HT + MSC animals compared to small focal
areas of cell loss in HT and MSC single treatments (arrows). (C) Injury scores were assessed in different brain regions (cortex, hippocampus, striatum, thalamus) resulting in a
sum score quantified for each animal (left column). Quantification of regional injury scores in the hippocampus is depicted in the right column. Bars present median values.
n = 14–16/group, *p < 0.05, **p < 0.01, ***p < 0.001, NT = normothermia, HT = hypothermia, MSC = mesenchymal stem cells.
124 J. Herz et al. / Brain, Behavior, and Immunity 70 (2018) 118–130

Fig. 3. Differential effects of acute hypothermia and delayed MSC therapy on grey and white matter injury. P9 mice were exposed to HI followed by 4 h NT or HT. MSC were
administered intranasally at P12. Protein lysates were isolated from ipsilateral hemispheres at the hippocampal (A, B) and the striatal level (C, D) at P16. Protein abundance of
MAP-2 and MBP was quantified by western blot. Data were normalized to the reference protein GAPDH and sham controls. n = 12–14/group, *p < 0.05, **p < 0.01, ***p < 0.001,
NT = normothermia, HT = hypothermia, MSC = mesenchymal stem cells.

therapy-effects were diminished resulting in no significant differ- results, HI-induced up-regulation of the endothelial adhesion
ences compared to NT animals (Fig. 4A, B). While neuronal density molecule VCAM-1 in NT animals was significantly reduced by HT
is decreased, HI induced a significant increase in oligodendrocytes and MSC (Fig. 5C), which was reversed after combining both ther-
in most severely affected brain regions (i.e. striatum, hippocam- apies (Fig. 5C). Whether increased VCAM-1 expression was caused
pus) which was not modulated by HT, MSC or combined treatment by general modulation of vascular integrity was examined by eval-
(Fig. 4C, D). uation of blood vessel density and diameters. HI-induced vessel
loss and dilation was improved by all therapeutic interventions
(Fig. 5D).
3.3. Anti-inflammatory effects of acute hypothermia and delayed MSC
treatment are reversed after combined treatment
3.4. MSC home to HI-injured brains independent of treatment
To dissect the underlying mechanisms of opposite effects by
the combined treatment, we analyzed sub-acute neuroinflamma- To investigate whether observed effects might have been
tory responses. HT, MSC and the combination of both demon- caused by impaired MSC homing in HT-treated animals we
strated a marked down-regulation of microglia activation quantified GFP-labelled MSC in brains of NT and HT-treated ani-
assessed by Iba-1 protein expression (Fig. 5A). As these results mals 14–16 h after intranasal delivery. These analyses revealed a
may not explain the detrimental interaction of both therapies, huge inter-individual variation, which was, however, independent
we analyzed peripheral leukocyte infiltration and endothelial acti- of treatment (Supplementary Fig. S5A,B) suggesting a similar
vation. Whereas HT and MSC inhibited HI-induced infiltration of migratory behavior of MSC in NT and HT animals. Of note, no
leukocytes, animals exposed to the combined treatment showed GFP-positive cells were detected in contralateral hemispheres
an increased leukocyte infiltration with no significant differences (Supplementary Fig. S5A). Overall cell number was low implicating
compared to normothermic controls (Fig. 5B). Similar to these that MSC may also have been distributed to other organ systems
J. Herz et al. / Brain, Behavior, and Immunity 70 (2018) 118–130 125

Fig. 4. Protection from regional neuronal loss by acute hypothermia and delayed MSC therapy is decreased after combining treatments. Regional neuronal and
oligodendrocyte densities were determined in cortex, striatum, hippocampus and thalamus of P16 mice exposed to HI followed by 4 h NT or HT on P9. MSC were
administered intranasally at P12. (A, B) Neuronal density was assessed by immunohistochemistry quantifying the NeuN-positive area. (C, D) Oligodendrocyte density was
quantified by counting Olig2-positive cells. Representative images in (A) and (C) show low and higher magnification images of the hippocampus. Scale bars: low
magnification: 250 mm, high magnification: 25 mm. n = 14–16/group, *p < 0.05, **p < 0.01, ***p < 0.001, NT = normothermia, HT = hypothermia, MSC = mesenchymal stem
cells.

thereby potentially mediating systemic effects. Therefore, we mea- TNFalpha and IL-10 while slightly increased levels of IL-4 in NT ani-
sured blood plasma cytokine levels for all experimental groups mals were significantly reduced by single MSC therapy and the
demonstrating no significant alteration by injury or therapy for combined treatment with HT (Supplementary Fig. S5C). However,
126 J. Herz et al. / Brain, Behavior, and Immunity 70 (2018) 118–130

Fig. 5. Anti-inflammatory effects of hypothermia and MSC on endothelial activation and leukocyte infiltration are reversed after combination therapy. P9 mice were exposed
to HI followed by 4 h NT or HT. MSC were administered intranasally at P12. Analysis was performed at P16. (A) Microglia activation was assessed via western blot quantifying
Iba-1 protein levels. (B) Leukocyte infiltration was evaluated by immunohistochemistry for CD45. Representative images show low and high magnification images of the
hippocampus. Scale bars: low magnification: 250 mm, high magnification: 25 mm. The CD45 positively stained area was quantified. (C) VCAM-1 expression was analyzed in
protein lysates via western blot. For (A) and (C) data were normalized to the reference protein GAPDH and sham controls. (D) Blood vessel density and diameter were
determined via immunohistochemistry. Mean and SD values of uninjured contralateral tissues are indicated by solid and dashed lines, respectively. n = 12–14/group for
(A) and (C), n = 14–16/group for (B) and (D), *p < 0.05, ***p < 0.001, NT = normothermia, HT = hypothermia, MSC = mesenchymal stem cells.

no differences were detected between MSC and HT + MSC treated inflammatory cytokine expression (e.g. IL-1beta and IL-6) while
animals, thus not explaining interaction effects for HI-induced expression of the anti-inflammatory cytokine IL-10 was reduced
brain injury. compared to co-culture with brain extracts derived from NT-
treated control mice (Fig. 6B). Furthermore, NT-brain homogenates
3.5. Modulation of MSC phenotype after culture with brain extracts of led to increased growth factor expression (e.g. BDNF and EGF)
hypothermia-treated animals which was significantly reduced after incubation with brain
extracts obtained from HT-treated animals (Fig. 6B). Analysis of
According to a similar homing of MSC to injured hemispheres in cell culture supernatants to determine protein secretion by MSC
NT and HT treated animals we hypothesized that MSC change their partially confirmed mRNA analysis demonstrating reduced levels
phenotype due to the changed in vivo microenvironment in cooled of IL-10 and IGF in HT-brain homogenate co-cultures compared
animals. Therefore, we analyzed gene expression in MSC for cytoki- to NT-brain co-cultures (Fig. 6C). For few analyses (e.g. BDNF and
nes and growth factors in MSC, co-cultured with conditioned med- TNFalpha) no significant differences were observed, most likely
ium containing brain extracts obtained from P12 mice exposed to due to the simultaneous time point of analysis, i.e. transcriptional
sham operation, HI + NT or HI + HT on P9 (Fig. 6A). In response to regulation preceding complex processes of protein translation and
culture with HT-brain homogenate, MSC up-regulated pro- intracellular targeting for extracellular secretion. Medium contain-
J. Herz et al. / Brain, Behavior, and Immunity 70 (2018) 118–130 127

Fig. 6. Increased pro-inflammatory cytokine expression and reduced growth factor expression by MSC exposed to brain extracts from hypothermia-treated animals. MSC
were cultured for 48 h with brain extracts obtained from P12 mice that were exposed to HI followed by 4 h NT or HT on P9 (A). mRNA expression (B) and protein analysis in
culture supernatants (C) for cytokines and growth factors was assessed by real time PCR on cell lysates (B) and ELISA/LuminexÒ in culture supernatants (C) in MSC co-cultures
with conditioned medium containing isolated brain extracts of normothermia (NT brain) or hypothermia (HT brain) treated animals (A). mRNA data were normalized to the
reference gene beta-2-mircoglobulin and to MSC co-cultured with brain homogenates of sham controls (n = 6, presented as dashed line, B). Dashed lines in (C) indicate
the mean value of MSC co-cultured with brain homogenates of sham controls (n = 4). n = 8/group for (B) and n = 6/group for (C). Data points reveal biological replicates/group,
*p < 0.05, **p < 0.01, ***p < 0.001 NT = normothermia, HT = hypothermia.

ing brain extracts at culture concentration (1 mg/ml) without cells treatment induced a further decline below control levels
demonstrated cytokine and growth factor concentrations below (Fig. 7B). Of note, VEGF is down-regulated by HI and improved
the detection limit, thus not confounding results of these analyses. by HT, MSC but also by the combination (Fig. 7B).

3.6. Combination of hypothermia and delayed MSC therapy increases 4. Discussion


pro-inflammatory cytokines and reduces neurotrophic factor levels in
HI-injured brains The present study shows that acute hypothermia (HT) following
neonatal hypoxia–ischemia (HI) results in protection of neuronal
To verify in vitro results cytokine and growth factor levels were structures and long-term motor outcome, while white matter
analyzed in brain tissue lysates 24 h after intranasal administra- injury and long-term cognitive function are not improved. In con-
tion of MSC or vehicle in NT and HT-treated animals. These analy- trast, a single delayed therapy with mesenchymal stem cells (MSC)
ses partially confirmed in vitro data, e.g. IL-6 and IL-1beta levels reduced HI-induced hypomyelination and memory deficits. Sur-
were increased by the combination of HT and MSC compared to prisingly, the combined treatment reversed protective effects of
single therapies (Fig. 7A). For IL-10 slightly increased levels in sin- each single therapy. Inhibitory effects on endothelial activation
gle treatments were reduced by HT + MSC though not reaching sig- and associated peripheral leukocyte infiltration by both monother-
nificance (Fig. 7A). Growth factor concentrations (except for VEGF) apies were abolished after combination. As a potential mechanism
were increased to abnormal high levels by HI but normalized to we provide evidence that modulation of the cerebral microenvi-
control levels by HT and MSC single therapy, while the combined ronment by HT alters the MSC phenotype, resulting in increased
128 J. Herz et al. / Brain, Behavior, and Immunity 70 (2018) 118–130

Fig. 7. Modulation of cytokine and growth factor levels by acute hypothermia and delayed intranasal MSC delivery 4 days after HI. P9 mice were exposed to HI followed by
4 h NT or HT. MSC were administered intranasally at P12. Cytokine (A) and growth factor (B) analysis was performed 24 h after MSC delivery. Protein lysates of ischemic
hemispheres were analyzed by LuminexÒ (TNFalpha, IL-1beta, IL-6, IL-10, VEGF) and ELISA (BDNF, EGF, IGF). Dashed lines indicate mean values of sham-operated control
mice (n = 7). n = 8/group, *p < 0.05, **p < 0.01, ***p < 0.001 NT = normothermia, HT = hypothermia, MSC = mesenchymal stem cells.

expression of pro-inflammatory cytokines and reduced expression therapies, we evaluated the underlying mechanisms instead of
of growth factors, leading to impaired restoration of long-term investigating further time points. MSC homing may have been
neurobehavioral development. impaired due to anti-inflammatory signals elicited by HT, resulting
Despite an increasing number of currently initiated clinical trials in reduced abundance of pro-inflammatory chemo-attractants for
of cell based therapies treating neonatal brain injury, only few pre- MSC migration to the injury site. However, the amount of accumu-
clinical reports focused on the combined treatment with HT (Fleiss lated MSC in injured hemispheres of NT and HT animals was sim-
et al., 2014). Intraventricular delivery of human umbilical cord ilar. Furthermore, our results revealed that both single treatment
blood MSC prior to HT revealed significant long-term protection effects were reversed, i.e. HT-induced protection is decreased after
from brain atrophy in a rodent model of neonatal HI (Park et al., additive MSC therapy. These results support the hypothesis that
2015). However, with regard to clinical translation less invasive MSC reach the injury region in HT mice, but their neuroprotective
delivery routes with a prolonged therapeutic window would be pre- capacity seems to be altered by acute HT preceding delayed MSC
ferred. Therefore, we applied MSC in the sub-acute phase through therapy.
the nasal route, reflecting a clinically applicable treatment design. Protective effects on microglia were similar for HT, MSC and the
Unexpectedly, we observed a negative interplay of both therapies. combined treatment. Furthermore, HI-induced downregulation of
Confirming previous reports (Burnsed et al., 2015; Reinboth VEGF is improved by all treatments at P13, closely resembling
et al., 2016; Sabir et al., 2012), we show that HT promotes short- effects on vessel density and integrity observed at P16. Therefore,
term protection, particularly from neuronal/axonal injury, result- detrimental interaction effects may be rather attributed to modu-
ing in long-lasting improvement of motor deficits. However, myeli- lation of endothelial activation and associated leukocyte infiltra-
nation was not restored by HT leading to impaired long-term tion. Inhibition of HI-induced up-regulation of vascular cell
cognitive abilities. MSC only, but not HT or HT + MSC therapy, adhesion molecule expression (as a marker of endothelial activa-
improved myelination and memory function. These results under- tion) and associated peripheral leukocyte infiltration by single
line the importance of white matter development for long-term therapies was absent in the combined treatment. These results
neurological outcome after perinatal brain injury. Nevertheless, suggest that MSC exposed to the anti-inflammatory environment
our results seem to contrast a previous report revealing that myeli- induced by HT, may increase inflammation. This is supported by
nation is enhanced by HT, which might be explained by different our in vitro analyses, demonstrating that MSC exposed to HT brain
methods of analyses (immunohistochemistry vs. western blot) extracts increase expression of pro-inflammatory cytokines (e.g.
and by the fact that protection was regionally restricted to the cau- IL-6 and IL-1beta) while reducing expression of the anti-
date nucleus (Koo et al., 2017). inflammatory cytokine IL-10. Similar results were obtained
Since in the investigated therapy design combined treatment in vivo revealing increased IL-6 and IL-1beta concentrations in
with MSC and HT revealed detrimental effects compared to single the combined treatment compared to single therapies early after
J. Herz et al. / Brain, Behavior, and Immunity 70 (2018) 118–130 129

MSC administration. This pro-inflammatory environment most deteriorated long-term protection from neurological deficits com-
likely contributed to induction of vascular cell adhesion molecule pared to single therapies. We provide evidence that the detrimen-
expression (O’Carroll et al., 2015) resulting in increased peripheral tal interaction may be most likely caused by an alteration of the
leukocyte infiltration. MSC phenotype in the HT-treated brain resulting in increased
In addition to immunomodulation, MSC act through promotion inflammatory responses and reduced expression of pro-
of regeneration via growth factor release. Importantly, MSC regenerative factors. Even though we confirmed MSC migration
expressed more BDNF and EGF following contact with an HI envi- into injured HI brains in rodents, further investigations especially
ronment in vitro, supporting previous reports that MSC promote a in higher developed organisms comparable to humans are needed
pro-regenerative environment in the HI brain (Donega et al., 2014; to elucidate the possible disposition of intranasally applied cells
van Velthoven et al., 2010b). However, after exposure to HT brain and their distribution within the brain to demonstrate safety and
extracts BDNF expression significantly decreased suggesting that efficacy of this approach. Furthermore, several risk factors con-
MSC’s pro-regenerative function is impaired in the brain of HT- tributing to neonatal encephalopathy should be considered. For
treated mice. Of note, in vivo analysis of BDNF, EGF and IGF demon- instance, in cases of infection-sensitized HI loss of protection by
strated an abnormal increase in HI-injured brains, which was HT (Osredkar et al., 2014) most likely caused by overwhelming
downregulated to control levels by single therapies while the com- inflammation might be overcome by stem cell therapy. Neverthe-
bined treatment induced a further decline below control levels. less, our unexpected results call for caution for precipitately
These results support the hypothesis that a certain threshold con- designed clinical trials for adjunctive cell-based therapies in com-
centration of these factors is particularly important for outcome, bination with hypothermia, since careful research is required to
with mild to moderate changes in growth factor signaling having define the optimum timing and application mode.
potentially positive effects, but extremes being unfavorable
(Binder et al., 2001; Chavko et al., 2002; Conover, 2016). Taken 5. Source of funding
together, our results suggest that HT modulates MSC function
inducing a pro-inflammatory environment combined with alter- This work was supported by the German Research Council (FE
ation of the homeostatic growth factor milieu which may account 518/5-1, UFM), an IFORES grant by the medical faculty (JH) and
for the observed reduction in therapeutic efficacy on myelination grants by the C.D. and the Karl-Heinz-Frenzen foundation
and long-term cognitive function. (UFM;IB).
Our study has strengths and limitations. Strengths of the
current study are large animal numbers and assessment of 6. Disclosure
long-lasting motor and also cognitive outcome, not previously
investigated. Multimodal approaches on potential underlying None
mechanisms were used to help to define more appropriate treat-
ment designs. We administered bone marrow derived MSC from Acknowledgment
the same species with well characterized treatment effects, a wide
therapeutic window, the possibility of a non-invasive application We thank R. Herrmann for assistance and discussion of real
mode combined with a reduced risk for adverse effects driven by time PCR data, S. Lupus for technical assistance in LuminexÒ
a potential mismatch between human and murine systems. A assays, J. Göthert and his team providing the opportunity to
potential limitation regarding comparability to previous work perform flow cytometry analysis with BD FACS LSRII.
and current initiated clinical trials is the use of bone marrow
MSC instead of cells derived from the umbilical cord, which are
Appendix A. Supplementary data
supposed to be more primitive than MSC isolated from adult tissue
sources. However, their therapeutic capacity is still under debate
Supplementary data associated with this article can be found, in
(Fleiss et al., 2014). The detected cell number of infiltrated MSC
the online version, at https://doi.org/10.1016/j.bbi.2018.02.006.
was low and revealed a huge inter-individual variation not allow-
ing reliable immunohistochemical analysis to determine spatial
distribution of cells. Even though this was independent of NT or References
HT treatment, further systematic kinetic analyses are needed to
determine the spatiotemporal distribution of MSC after intranasal Azzopardi, D., Strohm, B., Marlow, N., Brocklehurst, P., Deierl, A., Eddama, O.,
delivery. Another potential limitation might be that stereology Goodwin, J., Halliday, H.L., Juszczak, E., Kapellou, O., Levene, M., Linsell, L., Omar,
O., Thoresen, M., Tusor, N., Whitelaw, A., Edwards, A.D., Group, T.S., 2014. Effects
methods have not been applied. However, quantification was per- of hypothermia for perinatal asphyxia on childhood outcomes. N. Engl. J. Med.
formed according to our previous study (Reinboth et al., 2016) 371, 140–149
where we established the injury model and immunohistochemical Bevins, R.A., Besheer, J., 2006. Object recognition in rats and mice: a one-trial non-
matching-to-sample learning task to study ’recognition memory’. Nat. Protoc. 1,
analyses. When comparing different quantification methods (serial 1306–1311.
section and total region analysis vs. methods described here) a Binder, D.K., Croll, S.D., Gall, C.M., Scharfman, H.E., 2001. BDNF and epilepsy: too
close correlation was observed between both methods (not shown) much of a good thing? Trends Neurosci. 24, 47–53.
Burnsed, J.C., Chavez-Valdez, R., Hossain, M.S., Kesavan, K., Martin, L.J., Zhang, J.,
while the inter-individual, i.e. biological variability was high, as Northington, F.J., 2015. Hypoxia-ischemia and therapeutic hypothermia in the
frequently reported in this injury model (Engels et al., 2017; neonatal mouse brain–a longitudinal study. PLoS One 10, e0118889.
Patel et al., 2015; Rice et al., 1981; Sabir et al., 2012). Therefore, Chavko, M., Nadi, N.S., Keyser, D.O., 2002. Activation of BDNF mRNA and protein
after seizures in hyperbaric oxygen: implications for sensitization to seizures in
we choose to include higher biological replicates (i.e. sample size) re-exposures. Neurochem. Res. 27, 1649–1653.
rather than increasing technical replicates. Even though we applied Conover, C.A., 2016. Discrepancies in insulin-like growth factor signaling? No, not
a broad set of behavior tests covering locomotion, anxiety and really. Growth Horm. IGF Res. 30–31, 42–44.
Donega, V., Nijboer, C.H., van Tilborg, G., Dijkhuizen, R.M., Kavelaars, A., Heijnen, C.J.,
impulsive related behavior as well as cognitive function, further
2014. Intranasally administered mesenchymal stem cells promote a
cognitive tests, e.g. addressing learning behavior might be critical regenerative niche for repair of neonatal ischemic brain injury. Exp. Neurol.
to confirm our findings. 261, 53–64.
Our results confirm efficacy but also limitations of HT and MSC Donega, V., van Velthoven, C.T., Nijboer, C.H., van Bel, F., Kas, M.J., Kavelaars, A.,
Heijnen, C.J., 2013. Intranasal mesenchymal stem cell treatment for neonatal
monotherapy for treatment of neonatal HI. In contrast to our brain damage: long-term cognitive and sensorimotor improvement. PLoS One 8,
hypothesis, the combination of acute HT with delayed MSC therapy e51253.
130 J. Herz et al. / Brain, Behavior, and Immunity 70 (2018) 118–130

Engels, J., Elting, N., Braun, L., Bendix, I., Herz, J., Felderhoff-Muser, U., Dzietko, M., Shriver, N.N.R.N. Cognitive outcomes after neonatal encephalopathy.
2017. Sildenafil Enhances Quantity of Immature Neurons and Promotes Pediatrics 135, e624–634.
Functional Recovery in the Developing Ischemic Mouse Brain. Dev. Neurosci. Park, W.S., Sung, S.I., Ahn, S.Y., Yoo, H.S., Sung, D.K., Im, G.H., Choi, S.J., Chang, Y.S.,
39, 287–297. 2015. Hypothermia augments neuroprotective activity of mesenchymal stem
Eunson, P., 2015. The long-term health, social, and financial burden of hypoxic- cells for neonatal hypoxic-ischemic encephalopathy. PLoS One 10, e0120893.
ischaemic encephalopathy. Dev. Med. Child Neurol. 57 (Suppl 3), 48–50. Patel, S.D., Pierce, L., Ciardiello, A., Hutton, A., Paskewitz, S., Aronowitz, E., Voss, H.U.,
Ferriero, D.M., 2004. Neonatal brain injury. N. Engl. J. Med. 351, 1985–1995. Moore, H., Vannucci, S.J., 2015. Therapeutic hypothermia and hypoxia-ischemia
Fleiss, B., Guillot, P.V., Titomanlio, L., Baud, O., Hagberg, H., Gressens, P., 2014. Stem in the term-equivalent neonatal rat: characterization of a translational
cell therapy for neonatal brain injury. Clin. Perinatol. 41, 133–148. preclinical model. Pediatr. Res. 78, 264–271.
Gunn, A.J., Laptook, A.R., Robertson, N.J., Barks, J.D., Thoresen, M., Wassink, G., Reinboth, B.S., Koster, C., Abberger, H., Prager, S., Bendix, I., Felderhoff-Muser, U.,
Bennet, L., 2017. Therapeutic hypothermia translates from ancient history in to Herz, J., 2016. Endogenous hypothermic response to hypoxia reduces brain
practice. Pediatr. Res. 81, 202–209. injury: Implications for modeling hypoxic-ischemic encephalopathy and
Herz, J., Hagen, S.I., Bergmuller, E., Sabellek, P., Gothert, J.R., Buer, J., Hansen, W., therapeutic hypothermia in neonatal mice. Exp. Neurol. 283, 264–275.
Hermann, D.M., Doeppner, T.R., 2014. Exacerbation of ischemic brain injury in Rice 3rd, J.E., Vannucci, R.C., Brierley, J.B., 1981. The influence of immaturity on
hypercholesterolemic mice is associated with pronounced changes in hypoxic-ischemic brain damage in the rat. Ann. Neurol. 9, 131–141.
peripheral and cerebral immune responses. Neurobiol. Dis. 62, 456–468. Sabir, H., Scull-Brown, E., Liu, X., Thoresen, M., 2012. Immediate hypothermia is not
Herz, J., Sabellek, P., Lane, T.E., Gunzer, M., Hermann, D.M., Doeppner, T.R., 2015. neuroprotective after severe hypoxia-ischemia and is deleterious when delayed
Role of Neutrophils in Exacerbation of Brain Injury After Focal Cerebral Ischemia by 12 hours in neonatal rats. Stroke 43, 3364–3370.
in Hyperlipidemic Mice. Stroke 46, 2916–2925. Sheldon, R.A., Sedik, C., Ferriero, D.M., 1998. Strain-related brain injury in neonatal
Koo, E., Sheldon, R.A., Lee, B.S., Vexler, Z.S., Ferriero, D.M., 2017. Effects of mice subjected to hypoxia-ischemia. Brain Res. 810, 114–122.
therapeutic hypothermia on white matter injury from murine neonatal Thornton, C., Rousset, C.I., Kichev, A., Miyakuni, Y., Vontell, R., Baburamani, A.A.,
hypoxia-ischemia. Pediatr. Res. Fleiss, B., Gressens, P., Hagberg, H., 2012. Molecular mechanisms of neonatal
Lister, R.G., 1987. The use of a plus-maze to measure anxiety in the mouse. brain injury. Neurol. Res. Int. 2012, 506320.
Psychopharmacology 92, 180–185. van Velthoven, C.T., Kavelaars, A., van Bel, F., Heijnen, C.J., 2010a. Mesenchymal
Milner, L.C., Crabbe, J.C., 2008. Three murine anxiety models: results from multiple stem cell treatment after neonatal hypoxic-ischemic brain injury improves
inbred strain comparisons. Genes Brain Behav. 7, 496–505. behavioral outcome and induces neuronal and oligodendrocyte regeneration.
O’Carroll, S.J., Kho, D.T., Wiltshire, R., Nelson, V., Rotimi, O., Johnson, R., Angel, C.E., Brain Behav. Immun. 24, 387–393.
Graham, E.S., 2015. Pro-inflammatory TNFalpha and IL-1beta differentially van Velthoven, C.T., Kavelaars, A., van Bel, F., Heijnen, C.J., 2010b. Repeated
regulate the inflammatory phenotype of brain microvascular endothelial cells. J. mesenchymal stem cell treatment after neonatal hypoxia-ischemia has distinct
Neuroinflammation 12, 131. effects on formation and maturation of new neurons and oligodendrocytes
Osredkar, D., Thoresen, M., Maes, E., Flatebo, T., Elstad, M., Sabir, H., 2014. leading to restoration of damage, corticospinal motor tract activity, and
Hypothermia is not neuroprotective after infection-sensitized neonatal sensorimotor function. J. Neurosci. 30, 9603–9611.
hypoxic-ischemic brain injury. Resuscitation 85, 567–572. Walf, A.A., Frye, C.A., 2007. The use of the elevated plus maze as an assay of anxiety-
Pappas, A., Shankaran, S., McDonald, S.A., Vohr, B.R., Hintz, S.R., Ehrenkranz, R.A., related behavior in rodents. Nat. Protocols 2, 322–328.
Tyson, J.E., Yolton, K., Das, A., Bara, R., Hammond, J., Higgins, R.D., 2015. Wassink, G., Gunn, E.R., Drury, P.P., Bennet, L., Gunn, A.J., 2014. The mechanisms and
Hypothermia Extended Follow-up Subcommittee of the Eunice Kennedy treatment of asphyxial encephalopathy. Front. Neurosci. 8, 40.

You might also like