You are on page 1of 12

NEUROLOGICAL PROGRESS

Cell Therapy for Neonatal Hypoxia–


Ischemia and Cerebral Palsy
Laura Bennet, PhD,1 Sidhartha Tan, MD,2 Lotte Van den Heuij, MSc,1
Matthew Derrick, MBBS,2 Floris Groenendaal, MD, PhD,3 Frank van Bel, MD, PhD,3
Sandra Juul, MD, PhD,4 Stephen A. Back, MD, PhD,5 Frances Northington, MD, PhD,6
Nicola J. Robertson, PhD, FRCPCH,7 Carina Mallard, PhD,8
and Alistair Jan Gunn, MBChB, PhD1,9

Perinatal hypoxic–ischemic brain injury remains a major cause of cerebral palsy. Although therapeutic hypothermia is
now established to improve recovery from hypoxia–ischemia (HI) at term, many infants continue to survive with
disability, and hypothermia has not yet been tested in preterm infants. There is increasing evidence from in vitro and
in vivo preclinical studies that stem/progenitor cells may have multiple beneficial effects on outcome after hypoxic–
ischemic injury. Stem/progenitor cells have shown great promise in animal studies in decreasing neurological
impairment; however, the mechanisms of action of stem cells, and the optimal type, dose, and method of
administration remain surprisingly unclear, and some studies have found no benefit. Although cell-based
interventions after completion of the majority of secondary cell death appear to have potential to improve functional
outcome for neonates after HI, further rigorous testing in translational animal models is required before randomized
controlled trials should be considered.
ANN NEUROL 2012;71:589–600

Neonatal Encephalopathy at Term insults can be associated with transient recovery of oxida-
tive metabolism after reperfusion in a so-called latent
N eonatal encephalopathy due to perinatal hypoxia–
ischaemia (HI) occurs in 1 to 3 per 1,000 births at
1
term and is associated with high mortality and morbid-
phase (Fig 1).5 In this latent phase, the electroencephalo-
gram and cerebral blood flow are typically suppressed,
ity, with life-long chronic disabilities including cerebral but cerebral oxygenation is normal.6,7 After approxi-
palsy (CP).2,3 Neuropathological injury typically involves mately 6 to 15 hours, there may be secondary deteriora-
neuronal death in the cerebral cortex, hippocampus, and tion, with delayed seizures, progressive mitochondrial
basal ganglia–thalamus, with increasing evidence for failure,5,8 cytotoxic edema, and secondary cell death over
global white matter involvement.4 many days.6 The severity of this secondary deterioration
To interpret preclinical studies of neuroprotectants, is closely correlated with later neurodevelopmental
it is critical to appreciate why the latent (early recovery) impairment.9 Although considerable cell death typically
phase has been the effective therapeutic target for the occurs during the secondary phase, this is often followed
majority of potential treatments to date. Although a suf- by a chronic or tertiary phase of further progressive cell
ficiently severe/prolonged period of HI (the primary death, repair, and remodeling.10,11 It is believed that this
phase of injury) may be associated with immediate cell additional loss of neurons and glial cells is related to
death due to metabolic failure, even relatively severe chronic loss of trophic factors and synaptic input from

View this article online at wileyonlinelibrary.com. DOI: 10.1002/ana.22670

Received Jun 17, 2011, and in revised form Oct 1, 2011. Accepted for publication Nov 4, 2011.

Address correspondence to Dr Gunn, University of Auckland, Physiology and Paediatrics, Auckland, New Zealand. E-mail: aj.gunn@auckland.ac.nz

From the 1Department of Physiology, University of Auckland, Auckland, New Zealand; 2Department of Pediatrics, NorthShore University HealthSystem,
Evanston, IL; 3Department of Neonatology, University Medical Center Utrecht, Utrecht, the Netherlands; 4Department of Pediatrics, University of
Washington, Seattle, WA; 5Department of Pediatrics, Oregon Health and Science University, Portland, OR; 6Division of Neonatology and Neonatal Research
Laboratory, Johns Hopkins School of Medicine, Baltimore, MD; 7Institute for Women’s Health, University College London, London, UK; 8Institute for
Neuroscience and Physiology, Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; and 9Department of
Paediatrics, University of Auckland, Auckland, New Zealand.

V
C 2012 American Neurological Association 589
ANNALS of Neurology

HI or CP, 6 of them active, mostly using human umbili-


cal cord blood cells (hUCBCs). More than 200 patients
have been infused with hUCBCs for neonatal brain
injury and childhood motor deficits including CP, almost
entirely in uncontrolled studies (Fig 2).20 These data also
highlight the remarkable range of volumes given to
patients, consistent with the lack of knowledge of the
optimal dose of hUCBCs.
The safety of autologous cord blood cells appears
to be relatively good. A minor issue for late treatment is
that preserved cells are stored with dimethyl sulfoxide
(DMSO), and despite washing, thawed cells typically
FIGURE 1: Schematic representation of the evolution of include some residual DMSO, which can be associated
neural injury after exposure to hypoxia–ischemia. with acute adverse reactions.21 For example, in a pilot
study in children with acquired brain disorders aged 6
neighboring cells,12 and loss of or failure of recruitment days to 9.5 years, 76% of them with CP,22 3 of 184
of neural stem cells (NSCs) and glial progenitor cells patients developed anaphylactic reactions despite prophy-
within the brain.13 lactic antihistamines, bronchodilators, and steroids; all
In term infants with moderate to severe encephalop- adverse symptoms resolved rapidly after stopping the cell
athy, it is now well established that mild to moderate hypo- infusions and appropriate medical treatment.
thermia induced in the first 6 hours after birth, and contin- In contrast, although a detailed discussion is outside
ued for at least 72 hours, can significantly improve survival the scope of this paper, the potential for nonautologous
without disability.3,14 However, many infants still survive embryonic or pluripotent stem cells to differentiate into
with disability with current treatment protocols.3 Promising all cell types is associated with potential risk of terato-
adjunctive treatments including erythropoietin and xenon mas.23,24 Embryonic stem cells have been reported to
are now undergoing phase 2 clinical trial.15,16 In preterm form teratomas in mice of the same genetic back-
infants, the precise timing of neural injury is often unclear, ground,24 or in immunodeficient animals.23 In contrast,
and there are unresolved safety concerns around cooling.17 immunocompetent mice (or xenogenic rats) efficiently
Although there is increasing clinical evidence that maternal immunorejected the cells through a natural killer cell
treatment with magnesium sulfate before early preterm mechanism.25 These balanced risks of tumors and rapid
birth is associated with a small reduction in risk of CP in rejection need further detailed evaluation.
surviving infants,18 there is still considerable uncertainty.19
Thus, additional, late interventions in the tertiary phase in
both term and preterm infants might have potential to Analysis Strategy
decrease chronic cell loss and maldevelopment due to lack
For the present review, an international group of
of survival signals, and help repair.
researchers in perinatal neuroprotection have examined
The complex etiology of HI calls for a therapy that
intervenes on multiple levels, including promoting cell
survival, regeneration, and functional reorganization.
Over the past decade, the possibility of utilizing progeni-
tor or stem cells as a therapy for HI has gained increas-
ing support. There are many different types of stem cells,
derived from embryonic, fetal, placental, and adult tis-
sues. Their promise derives from the potential to replace
lost neurons, protect host cells, and promote their growth
and differentiation, as well as modulate the host immune
FIGURE 2: (A) Number of patients with newborn brain
response, all of which might decrease disability after HI. injury and children with motor deficits who received human
Parents of children with CP have been a driving force for umbilical cord blood cells (hUCBCs). The drop in 2010 coin-
the use of stem cells to treat HI brain injury. A handful cided with the beginning of a randomized clinical trial at
Duke that was responsible for the majority of hUCBC
of sensational cases of apparent recovery have been
administration. (B) The volume of hUCBCs given extends
reported in the lay press, leading to inflated expectations. over a wide range. The number of cells is not known. Data
Clinicaltrials.gov now lists 7 clinical trials for neonatal are derived from the Cord Blood Registry.20

590 Volume 71, No. 5


Bennet et al: Stem Cells and HI

the evidence for therapeutic potential for neonatal HI in transplanted 3 days after HI at P7, motor deficits were
term infants with different stem/progenitor cell prepara- reduced from 3 to 9 weeks of age.32 At 10 to 12 weeks,
tions, including NSCs, NSCs derived from fetal tissues, grafts could be identified in approximately 2=3 of animals,
mesenchymal stem cells (MSCs), multipotent astrocytic and showed a neurochemical profile broadly consistent
stem cells (MASCs), and multipotent adult-derived pro- with normal neocortex, although typical neocortical
genitor cells (MAPCs), as well as hUCBCs. MASCs are layers did not develop even for grafts integrated into the
a special type of astrocyte derived from the subependy- sensorimotor cortex, and the morphology of some phe-
mal zones of postnatal day 1 (P1) to P6 rats, which notypes was different from controls.
retain NSC-like properties, including the ability to form Park et al showed that NSCs transplanted 7 days
neurons as well as glia.26 MAPCs are pluripotent progen- before HI integrated and became quiescent, and did not
itor cells, typically isolated from the bone marrow of differentiate until they were subjected to HI.33 After HI,
adult rodent donors,27 that retain the ability to differen- NSCs migrated to the pemumbra of the infarct, where
tiate into neuronal lineages. Studies were searched for on the cells differentiated into neurons and oligodendro-
PubMed using the strategy of (perinatal brain injury OR cytes, suggesting that additional environmental cues are
(hypoxia–ischemia AND neonatal brain injury)) AND needed for these cells to migrate and differentiate.
(cell therapy OR progenitor cells OR cord blood cells). Unfortunately, the authors did not assess whether there
For each relevant paper identified in this way, we then was any functional benefit. Imitola et al demonstrated
performed a related search. The results are summarized injury-mediated homing along a stromal cell-derived fac-
in the Table. tor 1a (SDF-1a) gradient toward the site of injury.34
Supporting a critical role for this factor, NSCs expressed
the SDF receptor, CXC chemokine receptor 4, and in
Experimental HI
vitro cells increased migration and proliferation when
All of the studies that we identified used some modifi-
SDF-1a was added to culture.34
cation of the Vannucci model of unilateral carotid ar-
Similarly, intracerebral injection of MASCs har-
tery ligation followed by a period of moderate hypoxia
vested from mice (P1–P6) in P7 rats was associated with
in neonatal rats or mice.28 This functional paradigm of
diffuse migration and an astrocytic morphology in non-
perinatal injury is associated with infarction in the mid-
hypoxic controls, whereas after HI cells migrated toward
dle artery territory, typically with no overt cell death in
the area of injury, and some MASCs in the injured area
the hemisphere contralateral to ligation. Most rodents
developed neuronal or astrocytic phenotypes.35 In con-
are highly altricial. Although P7 in the rat or mouse trast, Sato et al found that the area of cortical infarction
was originally chosen because this is the age of peak was reduced when NSCs cultured from E14 fetal rats
brain growth, which occurs at term in humans,29 at P7 were transplanted together with chondroitinase ABC 24
rodents are just starting cortical myelination, compara- hours after HI in P7 rats, but not when NSCs were
ble with 34 weeks gestation in humans. On balance, administered alone.36 The authors postulated that coinfu-
cortical maturity of the rat has been estimated to be sion facilitated NSC migration and adherence, allowing
most comparable to the term human at P12 to P13.30 the transplanted NSCs to more efficiently support host
Thus, the studies analyzed here represent acute, well- cells with neurotrophins, or may have facilitated access of
defined perinatal HI that is most characteristic of brain neurotrophins to host neurons.
injury at term, but at a mildly preterm stage of brain NSCs derived from human embryonic stem cells
maturation. were also able to disperse, integrate, and differentiate
well when grafted into the forebrain of P7 rats 24 hours
NSCs after HI, and showed significant sprouting.37 There was
NSCs have been the cell type most commonly studied in upregulation of genes involved in neurogenesis, gliogene-
preclinical models of neonatal HI, because of their sis, and neurotrophic support, with increased microglia
potential to differentiate into glial and neuronal lineages, in the transplanted hemispheres. Four weeks after treat-
and even endothelial cells. In early experiments, cortical ment, there were small improvements in contralateral
grafts from fetal rats ((embryonic day) E13) were trans- limb use and rotarod performance.37 Similarly, intraven-
planted 7 days after HI at P7, and the animals were sac- tricular injection of mouse ESCs 2 to 3 days after HI in
rificed 2 to 6 weeks after transplantation.31 There was no P7 mice was associated with survival of transplanted cells
effect on the extent of brain damage, although >80% of expressing neural markers after 8 months of recovery,
grafts survived. In contrast, when a cell suspension from and improved escape latency on the Morris water maze
the cortical sensorimotor region of fetal rats (E16) was test at 2 and 8 months after HI.38

May 2012 591


592
TABLE : Outcome of Cell-Based Interventions in Neonatal HI
ANNALS

Cell Type Administration Species Hypoxia Engraftment, Days Cellular/Molecular Outcome Reference

NSCs
NSCs Seeded onto a polymer P7 mice 120 min Appeared to fill area Donor cells on Some evidence Park 200239
of Neurology

scaffold, then implanted of infarction, with polymer scaffold of axonal


in cortex 7 days after HI vascularization differentiated into connection with
at 14 days. neurons and intact cortex.
oligodendrocytes. Reduced
astrogliosis and
inflammation.
NSCs transduced 3.0  105 cells in 8ll, P7 mice 120–180 Donor-derived Donor cells No functional Park 200676
with a retrovirus intracerebral, 3 days min neurons increased differentiated into outcome.
encoding NT-3 after HI from 5% to >80% cholinergic,
around the area of GABAergic, and
infarction, and 20% in glutamatergic
the infarct at 2–4 weeks. neurons.
NSCs derived 3  104 cells in 3ll, P7 mice 90 min Few cells in hippocampus Co-staining of Memory and Ma 200738
from mESC i.v., 2–3 days after HI and cerebral cortex at ESCs and NF. learning
2 and 8 months. improvement
after 2 and
8 months.
NSCs derived 3 injections of 1  106 P7 rats 90 min Engraftment at 4 weeks. Transplanted No change in Daadi 201037
from hESC cells in 2ll, cells ! NeuN, lesion size.
intrahemispheric, TuJ1, and GAD Improved use of
24 hours after HI expression, contralateral
enhanced axonal forelimb
sprouting. Increased and ameliorated
upregulation of locomotor deficits.
genes involved in
regeneration.
Rat neural 2.5  105 cells in P7 rats 120 min Some cells found N/A. Decreased lesion Sato 200836
progenitor cells 5ll, i.v., 24 hours in/around lesion at size and increased
from embryonic after HI 7 days. wet weight of brain.
forebrain
(with ChABC)

Volume 71, No. 5


TABLE (Continued)

Cell Type Administration Species Hypoxia Engraftment, Days Cellular/Molecular Outcome Reference

May 2012
Fetal cortical Fetal neocortical P7–P8 120–150 80% graft survival N/A. No decrease Elsayed 199631
grafts transplants from rats min rate, evidence of in lesion size.
E13 rat embryos, integration at 2
7 days after HI and 6 weeks.
Fetal cortical 1  104 cells in P7 rats 150 min Grafts were N/A. Significant Jansen 199732
grafts 2ll, sensorimotor identifiable improvement
cortex, 3 days after HI and integrated in motor tests
at 21 and 29 days. from P21 to P63.
Human fetal 5  105 in 5ll, left P7 rats 90 min Human cells present The majority of No functional Qu 200577
NSCs cerebral ventricle, in SVZ, corpus transplanted cells outcome.
3 days after HI callosum, cortex, differentiated into
and hippocampus, neurons and
particularly on astrocytes.
injured side.
MSCs
Rat MSCs 4  106 in 0.5ml, P7 rats 120 min MSCs present A few MSCs No functional Guan 200446
(derived intraperitoneal, throughout expressed nestin outcome.
from bone 2 hours after HI cerebra in both protein; no
marrow) groups at 14 days. differentiation
into neurons or
astrocytes.
hMSCs derived 1  106 cells in 100ll, P7 rats 210 min Cells found equally No change in Lee 201045
from bone marrow intracardiac, distributed in both hemispheric
3 days after HI hemispheres at 6 weeks. volume, but
improved
neurologic
performance.
Murine MSCs 1  105 cells in 2ll, P9 mice 45 min <1% of BrdU-positive Reduced neuronal Improved van Velthoven
(derived from intrahemispheric, cells were donor cells. and oligodendrocyte motor skills 201042
bone marrow) 3 or 10 days after HI loss 21 days after at P19 and P30.
HI, greater BrdU
incorporation in
the ischemic
hemisphere.
Bennet et al: Stem Cells and HI

593
594
TABLE (Continued)
ANNALS

Cell Type Administration Species Hypoxia Engraftment, Days Cellular/Molecular Outcome Reference

Murine MSCs 1  105 cells in 2ll, P9 mice 45 min Very few cells present in : host cell Reduced lesion van Velthoven
intrahemispheric, the granule cell layer of proliferation and size. Improved 201043
3 or 10 days after HI the dentate gyrus at differentiation motor skills
6 weeks. toward neurons P20-18-27.
of Neurology

and oligodendrocytes.
: immune cell
proliferation.
Murine MSCs 5  105 cells, P9 mice 45 min MSCs present in the Cocultured MSC þ ; gray and van Velthoven
intranasal, 10 days affected hemisphere brain extracts 10 white matter 201044
after HI but had not differentiated. days after HI, : loss.
CXCR4, FGF2,
SDF-1, NGF, and
NT; ; IL1b and
TGFb1 mRNA.
MASCs
mMASCs 5  103 cells in 1ll, P7 rats 120 min Cells found in lesion at Transplanted cells in No functional Zheng
somatosensory cortex, 3–21 days later. nonischemic brain outcome. 200635
24 hours and 5 days had astrocytic
after HI phenotype; a few
with neuronal
appearance in
injured cortex.
MAPCs
rMAPCs, derived 2  105 in 3ll, P7 rats 150 min Donor cells present in ; motor asymmetry Yasuhara
from bone marrow intrahippocampal, the hippocampal region. at P21 but no 200640
þ cyclosporin 7 days after HI difference in
rotarod test.
rMAPCs þ 2  105 cells in 3ll, P7 rats 150 min 0.18% of i.v., 0.51% Reduced neuronal ; motor asymmetry Yasuhara
cyclosporin i.v. (jugular) and of intracranial cells found loss in CA3 and : rotarod times 200841
intrahippocampal, in the CA3, CA2 of region of at P21 but not P14
7 days after HI hippocampus 14 hippocampus. after both i.v. and
days later. intracerebral
administration.

Volume 71, No. 5


TABLE (Continued)

May 2012
Cell Type Administration Species Hypoxia Engraftment, Days Cellular/Molecular Outcome Reference

hUCBCs
Human cord blood 1  107 cells in P7 rats 80 min Many cells in ischemic N/A. Normalized 12% Meier
mononuclear cells 500ll, intraperitoneal, hemisphere, 21 days. difference in toe 200647
24 hours after HI distances, 8%
step lengths.
Human cord blood 1  107 cells in P7 rats 120 min Few hUCB cells located No change in No effect on de Paula
mononuclear cells 100lL, i.v. (jugular), in rat brain, 21 days. volume of injured spatial memory 200951
24 hours after HI hemisphere after deficits.
21 days.
hUCBC-derived 1  105 cells in 2ll, P7 rats 150 min Differentiation into Apparent (subjective) Improved Xia 201078
mononuclear cells intracortical, astrocytes but not reduction in cortical neurological
(passage 10) þ 3 days after HI neurons, 7 days after neuronal loss at score, P24 to P38.
cyclosporine A transplant. 28 days.
Human cord blood 2  106 cells in P7 rats 90 min Few cells in ischemic Decreased cell Improved neonatal Pimentel-Coelho
mononuclear cells 200ll, intraperitoneal, cortex and striatum, death and reflexes at P11 201050
3 hours after HI 2 days. microglial but not at P14.
activation.
Human cord blood 1.5  104 cells in P7 rats 150 min Few cells in ischemic Increased growth 20–25% Yasuhara
mononuclear cells þ 200ll, i.v., hippocampus, 14 days. factors in brain, improvement 201049
mannitol 2–3 hours after HI CA1 dendrites. rotarod and
elevated body
swing tests at p14
and P21.
Human cord blood 1  107 cells in 500l, P7 rats 80 min Many cells in peri-infarct No change Improved Geissler
mononuclear cells intraperitoneal, area after 42 days. in size of sensorimotor 201148
24 hours after HI hemispheric lesion. function, cortical
maps, and receptive
fields, and reduced
hyperexcitability,
at P48.
BrdU ¼ bromodeoxyuridine; ChABC ¼ chondroitinase ABC; CXCR ¼ chemokine (C-X-C motif ) receptor; ESC ¼ embryonic stem cell; FGF ¼ fibroblast growth factor; GABA ¼
gamma-aminobutyric acid; GAD ¼ glutamate decarboxylase; h ¼ human; HI ¼ hypoxia–ischemia; i.v. ¼ intravenous; IL ¼ interleukin; MAPC ¼ multipotent adult-derived progeni-
tor cell; MASC ¼ multipotent astrocytic stem cell; MSC ¼ mesenchymal stem cell; N/A ¼ not applicable; NF ¼ neurofilament; NGF ¼ nerve growth factor; NSC ¼ neural stem
cell; NT ¼ neurotrophin; P ¼ postnatal day; SDF ¼ stromal cell-derived factor; SVZ ¼ subventricular zone; TGF ¼ transforming growth factor; Tuj1 ¼ Neuron-specific class III
beta-tubulin; UCBC ¼ umbilical cord blood cell.
Bennet et al: Stem Cells and HI

595
ANNALS of Neurology

In practical terms, these studies still represent com- 14 to 40 days after treatment despite no significant
paratively early treatment. Intriguingly, there is early evi- reduction in the volume of infarction.45 MSCs were
dence from the P7 rat that if NSCs are seeded onto a found in control brains and diffusely throughout the
polymer scaffold platform before transplantation, they lesioned and nonlesioned hemispheres; however, more
may be able to bridge evolving cystic lesions 7 days after cellular differentiation was seen after HI than sham HI.
HI and establish at least some connections to host tis- Similarly, Guan et al reported that MSCs were found
sue.39 Given the results of other studies, discussed below, extensively throughout the brain 14 days after intraperi-
where cells were given at least as late, it is unclear toneal infusion of MSCs 2 hours after HI in P7 rats.46
whether such a technological assist is needed. MSCs were increased in the injured hemisphere after HI
but showed little differentiation into neural cell types.
MAPCs
MAPCs given to P7 rat pups either into the hippocam- hUCBCs
pus or intravenously (i.v.) 7 days after HI were associated Finally, there is also evidence for benefit with hUCBCs
with a significant reduction in motor asymmetry 14 days in rodents. Meier et al reported that intraperitoneal injec-
after injection compared to controls.40,41 In the earlier tion of 107 hUCBCs 24 hours after HI in P7 rats was
study, intracerebral MAPC injection did not significantly associated with reduced spastic paresis despite no change
improve rotarod times (a measure of sensorimotor func- in the area of hemispheric infarction.47 The cells
tion),40 whereas the later study suggested a small migrated to the injured hemisphere within 3 days, but
improvement in rotarod times 14 days (ie, P21) after with no evidence of differentiation 14 days after adminis-
both intracerebral and i.v. administration. A potential tration. More recently, this group showed in the same
complicating aspect of this protocol is that all animals paradigm that similarly to their first report, hUCBCs
were treated with daily immunosuppression throughout given 24 hours after HI were associated with no effect on
the survival period. However, controls also received this the size of the lesion, and yet significantly preserved
intervention. MAPCs were present in hippocampus areas somatosensory function on the side of the lesion.48 Nota-
after both intracerebral and i.v. injection 14 days after bly, they found reduced hyperexcitability, and restoration
treatment (P21),40,41 with significant improvement in of the cortical maps and receptive fields in the somato-
numbers of CA3 neurons at P21 with both routes of sensory cortex. This strongly suggests that benefit was
administration.41 mediated by improved function of surviving brain tissue.
Similarly, Yasuhara et al found that i.v. administra-
MSCs tion of 1.5  104 hUCBCs, with or without cotreatment
Intracranial injection of mouse MSCs 3 days after HI in with mannitol, 7 days after HI in P7 rats was associated
P9 mice was associated with a 42% and 31% reduction with significant improvement in motor asymmetry and
in neuronal and oligodendrocyte loss, respectively, 28 coordination 7 and 14 days later.49 In this study, only a
days after HI.42 There was greater proliferation of endog- few cells were found in the damaged area. The authors
enous neural and glial cells, with reduced proliferation of suggested a paracrine effect as the levels of neural growth
microglia. Functional and histological improvement was factor, glial cell line-derived neurotrophic factor, and
seen when MSC administration was delayed until 10 brain-derived neurotrophic factor were increased 3 days
days after HI.42 Further functional improvements were after treatment, particularly when cells were combined
associated with repeated injections of MSCs both 3 and with mannitol to increase blood–brain barrier
10 days after HI.43 Finally, this group has presented evi- permeability.49
dence that in the same paradigm, MSCs given intrana- Consistent with these findings, Pimentel-Coelho et
sally 10 days after HI can improve outcome.44 Consist- al found that intraperitoneal injection of hUCBCs in P7
ent with the reports from other studies discussed above rats 3 hours after HI was associated with only trivial
that the benefit of cell treatment may not be due to new numbers of hUCBCs in the lesion, a small reduction in
cell formation, MSCs were present in the affected hemi- neuronal loss in the striatum, and small improvements in
sphere 28 days after HI, but they remained 2 sensorimotor reflexes 4 days after HI, but no signifi-
undifferentiated. cant difference from vehicle controls at 2 and 7 days.50
Further supporting the potential for systemic treat- The improved outcomes were associated with reduced
ment to improve functional outcomes, intracardiac injec- caspase-3 cleavage around the lesion site and an overall
tion of hMSCs 3 days after HI in P7 rats was associated reduction in activated microglia and macrophages con-
with significant improvement in 1 of 2 neuromotor tests sistent with antiapoptotic and anti-inflammatory actions.

596 Volume 71, No. 5


Bennet et al: Stem Cells and HI

However, in contrast, de Paula et el found that intrave- ygen free radicals as well as diminishing the infiltration
nous injection of hUCBCs 24 hours after HI in P7 rats of leukocytes.58 For example, multipotent progenitors
was associated with no significant improvement in either derived from human amnion can reduce inflammation,
volume of infarction or in spatial memory 3 weeks with decreased monocyte chemoattractant protein-1,
later.51 transforming growth factor (TNF)a, interleukin (IL)-1 /-
6, and profibrotic TNF-b expression and downregulation
of tissue inhibitors of matrix metalloproteinase-1 and -
Knowledge Gaps
2.65 These cells can attenuate white matter injury and
Although the optimal dose, timing, and mechanisms of
increase plasma concentrations of the anti-inflammatory
action for stem cells in animal models are still not clear
cytokine IL-10 in preterm fetal sheep after exposure to
from the studies above, they do provide reasonable evi-
lipopolysaccharide.66,67
dence that administration of progenitor/stem cells may
Neurotrophic factors are secreted by progenitor cells
have beneficial effects on histological and behavioral out-
in vitro, including neurotrophin-3, brain-derived neuro-
comes after neonatal HI. It is striking that these benefits
trophic factor, and glial-derived neurotrophic factor,
seem to be comparable between very distinct types of
which support and drive endogenous cell migration, pro-
stem cells, although some studies of similar cell types,
liferation, and differentiation.68 In the adult rat, Borlon-
timing, and routes of administration found either no or
gan et al reported that systemic injection of hUCBCs
restricted/conditional benefits.31,45,51 In practical terms,
improved behavioral outcome and lesion size in associa-
autologous hUCBCs have the major advantage of not
tion with increased neurotrophin levels in the brain.60
requiring immunosuppression,22,52 but this is balanced
Even when intravenous cell injection was combined with
by the hematogenous origin of the cells, and thus less
a blood–brain barrier permeabilization agent, no cells
clear commitment to neural differentiation.
engrafted. Similarly, even intracerebral injection of MSCs
in the neonatal rat was associated with trivial survival of
Mechanisms of Action injected MSCs (1% after 18 days),54 but marked
Early studies aimed to establish whether the cells could induction of genes supporting growth and survival in the
engraft and replace dying cells. Although in many studies brain.
the cells did survive and proliferate in the brain, the net
increase in cell numbers was generally negligible. Even Brain Injury Associated with Premature Birth
where differentiation was seen, it is not clear that it Premature birth is associated with a very high burden of
resulted in fully functional and integrated neurons or neurological injury, including a 15% risk of severe neuro-
glia. It is striking that more recent studies suggest little logical impairment, and up to 50% risk of cognitive defi-
differentiation,53 raising the possibility of methodological cits in later childhood.69 Although HI likely contributes
issues with cell identification in earlier studies. Much to at least some brain injury, chronic inflammation and
more importantly, this suggests that the key mechanisms many other poorly defined factors are involved,13 the
are actually release of neurotrophic and immunomodula- timing remains unclear, and preterm infants most often
tory/immune suppressive factors. develop diffuse white matter injury in the first few weeks
Consistent with this hypothesis, recent studies have rather than acute gray matter loss.70,71 As previously
confirmed significant functional improvements with pe- reviewed, this is followed by long-term impairment of
ripheral administration after neonatal HI, despite little the development of both gray matter and white matter
evidence of functional engraftment.41,44,45,54 Evidence structures.13
from adult models of stroke and central nervous system There is increasing preclinical evidence that exoge-
disease support the concept that progenitor/stem cells nous oligodendrocyte progenitor cells and glial-restricted
can attenuate tertiary damage associated with postisch- precursor cells can augment recovery from adult spinal
emic inflammation and secondary cell death due to lack cord injury, by augmenting the key supporting cells,
of environmental stimulation.55–61 Nevertheless, again, astrocytes, and oligodendrocytes.72 Given that classic
not all studies support functional benefits.51,62–64 periventricular white matter injury in preterm infants is
Treatment at different time points during the evolu- associated with axonopathy and depletion of oligoden-
tion of injury may allow a range of potentially comple- drocytes,73 these types of cells may be particularly helpful
mentary mechanisms to be targeted. If stem cells are for promoting repair of the preterm brain. However, we
administered relatively soon after HI, then many cells were unable to identify any relevant studies of perinatal
may be saved by attenuation of the immune response, brain injury that used these cells. Conceptually, it is im-
decreasing the release of excitotoxins, cytotoxins, and ox- portant to appreciate that over the past few decades the

May 2012 597


ANNALS of Neurology

prevalence of necrotic periventricular leukomalacia has 4. Gunn AJ, Bennet L. Fetal hypoxia insults and patterns of brain
injury: insights from animal models. Clin Perinatol 2009;36:579–593.
steadily decreased. Although subtle, astrogliotic lesions
5. Bennet L, Roelfsema V, Pathipati P, et al. Relationship between
remain common, and there is evidence that the myelin evolving epileptiform activity and delayed loss of mitochondrial
deficit associated with these milder lesions may be associ- activity after asphyxia measured by near-infrared spectroscopy in
preterm fetal sheep. J Physiol 2006;572:141–154.
ated with maturation arrest of oligodendrocyte precur-
sors, without axonopathy.74 The potential of these oligo- 6. Gunn AJ, Gunn TR, de Haan HH, et al. Dramatic neuronal rescue
with prolonged selective head cooling after ischemia in fetal
dendrocyte progenitors to myelinate is presently unclear. lambs. J Clin Invest 1997;99:248–256.
Thus, it will be critical to test whether cell-based inter- 7. Jensen EC, Bennet L, Hunter CJ, et al. Post-hypoxic hypoperfu-
ventions can promote normal white matter maturation sion is associated with suppression of cerebral metabolism and
increased tissue oxygenation in near-term fetal sheep. J Physiol
and myelination in white matter lesions. 2006;572:131–139.

8. Lorek A, Takei Y, Cady EB, et al. Delayed (‘‘secondary’’) cerebral


Conclusions energy failure after acute hypoxia-ischemia in the newborn piglet:
continuous 48-hour studies by phosphorus magnetic resonance
All of the studies reported to date of cell administration for spectroscopy. Pediatr Res 1994;36:699–706.
neonatal HI have been performed in rodents, which are a 9. Roth SC, Baudin J, Cady E, et al. Relation of deranged neonatal cerebral
poor model for human brain development. Laboratory oxidative metabolism with neurodevelopmental outcome and head cir-
cumference at 4 years. Dev Med Child Neurol 1997;39:718–725.
rodents are highly altricial, and their brains are much sim-
pler (lissencephalic), with very small white matter volumes 10. Colbourne F, Corbett D. Delayed postischemic hypothermia: a six
month survival study using behavioral and histological assess-
compared with humans.75 Based on previous experience, ments of neuroprotection. J Neurosci 1995;15:7250–7260.
this study group strongly recommends that efficacy must be 11. Geddes R, Vannucci RC, Vannucci SJ. Delayed cerebral atrophy
demonstrated in at least 2 nonrodent species with complex, following moderate hypoxia-ischemia in the immature rat. Dev
Neurosci 2001;23:180–185.
humanlike brains before undertaking definitive, large
12. Stone BS, Zhang J, Mack DW, et al. Delayed neural network
randomized controlled trials of cell-based interventions. In degeneration after neonatal hypoxia-ischemia. Ann Neurol 2008;
contrast, fresh autologous volume and red blood cell- 64:535–546.
reduced cord blood cells appear to be of sufficiently low 13. Barrett RD, Bennet L, Davidson J, et al. Destruction and recon-
risk to justify early safety and feasibility studies. Although struction: hypoxia and the developing brain. Birth Defects Res C
Embryo Today 2007;81:163–176.
there appears to be strong potential for cell-based interven-
14. Gunn AJ, Thoresen M. Hypothermic neuroprotection. NeuroRx
tions after neonatal HI, there is still much work to be done. 2006;3:154–169.
Clinically, a minimally invasive strategy with cells that do 15. Imperial College London. Neuroprotective effects of hypothermia
not raise ethical issues is essential. We need to determine combined with inhaled xenon following perinatal asphyxia (TOBYXe).
the most appropriate cell type, dose, mode, and timing of Study NCT00934700, 2009. Available at: http://clinicaltrials.gov
(accessed on 28 September 2011).
administration, as well as potential long-term side effects,
16. University of California, San Francisco. Neonatal erythropoietin in
to ensure safe translation into the clinical realm. asphyxiated term newborns (NEAT). Study NCT00719407, 2011. Avail-
able at: http://clinicaltrials.gov (accessed on 28 September 2011).
17. Gunn AJ, Bennet L. Brain cooling for preterm infants. Clin Perina-
Acknowledgment tol 2008;35:735–748.
L.B. and A.J.G. were supported by the Health Research 18. Committee Opinion No. 455: Magnesium sulfate before antici-
Council of New Zealand grants 11-576 and 09-065. pated preterm birth for neuroprotection. Obstet Gynecol 2010;
115:669–671.
19. Huusom LD, Secher NJ, Pryds O, et al. Antenatal magnesium sul-
Potential Conflicts of Interest phate may prevent cerebral palsy in preterm infants—but are we
Nothing to report. convinced? Evaluation of an apparently conclusive meta-analysis
with trial sequential analysis. BJOG 2011;118:1–5.

20. Cord Blood Registry. 2011. Available at: http://www.cordblood.


com/ (accessed on 16 April 2011).
References 21. Benekli M, Anderson B, Wentling D, et al. Severe respiratory
1. Kurinczuk JJ, White-Koning M, Badawi N. Epidemiology of neona- depression after dimethylsulphoxide-containing autologous stem
tal encephalopathy and hypoxic-ischaemic encephalopathy. Early cell infusion in a patient with AL amyloidosis. Bone Marrow Trans-
Hum Dev 2010;86:329–338. plant 2000;25:1299–1301.
2. Marlow N, Rose AS, Rands CE, Draper ES. Neuropsychological 22. Sun J, Allison J, McLaughlin C, et al. Differences in quality
and educational problems at school age associated with neonatal between privately and publicly banked umbilical cord blood units:
encephalopathy. Arch Dis Child Fetal Neonatal Ed 2005;90: a pilot study of autologous cord blood infusion in children with
F380–F387. acquired neurologic disorders. Transfusion 2010;50:1980–1987.
3. Edwards AD, Brocklehurst P, Gunn AJ, et al. Neurological out- 23. Dressel R, Schindehutte J, Kuhlmann T, et al. The tumorigenicity
comes at 18 months of age after moderate hypothermia for peri- of mouse embryonic stem cells and in vitro differentiated neuronal
natal hypoxic ischaemic encephalopathy: synthesis and meta- cells is controlled by the recipients’ immune response. PloS One
analysis of trial data. Br Med J 2010;340:c363. 2008;3:e2622.

598 Volume 71, No. 5


Bennet et al: Stem Cells and HI

24. Zhao T, Zhang ZN, Rong Z, Xu Y. Immunogenicity of induced plu- 43. van Velthoven CT, Kavelaars A, van Bel F, Heijnen CJ. Repeated
ripotent stem cells. Nature 2011;474:212–215. mesenchymal stem cell treatment after neonatal hypoxia-ischemia
has distinct effects on formation and maturation of new neurons
25. Dressel R, Nolte J, Elsner L, et al. Pluripotent stem cells are highly
and oligodendrocytes leading to restoration of damage, cortico-
susceptible targets for syngeneic, allogeneic, and xenogeneic nat-
spinal motor tract activity, and sensorimotor function. J Neurosci
ural killer cells. FASEB J 2010;24:2164–2177.
2010;30:9603–9611.
26. Laywell ED, Rakic P, Kukekov VG, et al. Identification of a multipo-
44. van Velthoven CT, Kavelaars A, van Bel F, Heijnen CJ. Nasal
tent astrocytic stem cell in the immature and adult mouse brain.
administration of stem cells: a promising novel route to treat neo-
Proc Natl Acad Sci U S A 2000;97:13883–13888.
natal ischemic brain damage. Pediatr Res 2010;68:419–422.
27. Keene CD, Ortiz-Gonzalez XR, Jiang Y, et al. Neural differentiation 45. Lee JA, Kim BI, Jo CH, et al. Mesenchymal stem-cell transplanta-
and incorporation of bone marrow-derived multipotent adult pro- tion for hypoxic-ischemic brain injury in neonatal rat model.
genitor cells after single cell transplantation into blastocyst stage Pediatr Res 2010;67:42–46.
mouse embryos. Cell Transplant 2003;12:201–213.
46. Guan XQ, Yu JL, Li LQ, Liu GX. Study on mesenchymal stem cells
28. Rice JE III, Vannucci RC, Brierley JB. The influence of immaturity entering the brain through the blood-brain barrier [in Chinese].
on hypoxic-ischemic brain damage in the rat. Ann Neurol 1981;9: Zhonghua Er Ke Za Zhi 2004;42:920–923.
131–141.
47. Meier C, Middelanis J, Wasielewski B, et al. Spastic paresis after
29. Dobbing J, Sands J. Comparative aspects of the brain growth perinatal brain damage in rats is reduced by human cord blood
spurt. Early Hum Dev 1979;3:79–83. mononuclear cells. Pediatr Res 2006;59:244–249.
30. Romijn HJ, Hofman MA, Gramsbergen A. At what age is the 48. Geissler M, Dinse HR, Neuhoff S, et al. Human umbilical cord
developing cerebral cortex of the rat comparable to that of the blood cells restore brain damage induced changes in rat somato-
full-term newborn human baby? Early Hum Dev 1991;26:61–67. sensory cortex. PloS One 2011;6:e20194.
31. Elsayed MH, Hogan TP, Shaw PL, Castro AJ. Use of fetal cortical 49. Yasuhara T, Hara K, Maki M, et al. Mannitol facilitates neurotro-
grafts in hypoxic-ischemic brain injury in neonatal rats. Exp Neurol phic factor up-regulation and behavioural recovery in neonatal
1996;137:127–141. hypoxic-ischaemic rats with human umbilical cord blood grafts. J
32. Jansen EM, Solberg L, Underhill S, et al. Transplantation of fetal Cell Mol Med 2010;14:914–921.
neocortex ameliorates sensorimotor and locomotor deficits follow- 50. Pimentel-Coelho PM, Magalhaes ES, Lopes LM, et al. Human cord
ing neonatal ischemic-hypoxic brain injury in rats. Exp Neurol blood transplantation in a neonatal rat model of hypoxic-ischemic
1997;147:487–497. brain damage: functional outcome related to neuroprotection in
33. Park KI, Hack MA, Ourednik J, et al. Acute injury directs the the striatum. Stem Cells Dev 2010;19:351–358.
migration, proliferation, and differentiation of solid organ stem 51. de Paula S, Vitola AS, Greggio S, et al. Hemispheric brain injury
cells: evidence from the effect of hypoxia-ischemia in the CNS on and behavioral deficits induced by severe neonatal hypoxia-ische-
clonal ‘‘reporter’’ neural stem cells. Exp Neurol 2006;199:156–178. mia in rats are not attenuated by intravenous administration of
34. Imitola J, Raddassi K, Park KI, et al. Directed migration of neural human umbilical cord blood cells. Pediatr Res 2009;65:631–635.
stem cells to sites of CNS injury by the stromal cell-derived factor 52. Escolar ML, Poe MD, Provenzale JM, et al. Transplantation of um-
1alpha/CXC chemokine receptor 4 pathway. Proc Natl Acad Sci bilical-cord blood in babies with infantile Krabbe’s disease. N
U S A 2004;101:18117–18122. Engl J Med 2005;352:2069–2081.
35. Zheng T, Rossignol C, Leibovici A, et al. Transplantation of multi- 53. Rizvanov AA, Kiyasov AP, Gaziziov IM, et al. Human umbilical
potent astrocytic stem cells into a rat model of neonatal hypoxic- cord blood cells transfected with VEGF and L(1)CAM do not dif-
ischemic encephalopathy. Brain Res 2006;1112:99–105. ferentiate into neurons but transform into vascular endothelial
cells and secrete neuro-trophic factors to support neuro-genesis—
36. Sato Y, Nakanishi K, Hayakawa M, et al. Reduction of brain injury
a novel approach in stem cell therapy. Neurochem Int 2008;53:
in neonatal hypoxic-ischemic rats by intracerebroventricular injec-
389–394.
tion of neural stem/progenitor cells together with chondroitinase
ABC. Reprod Sci 2008;15:613–620. 54. van Velthoven CT, Kavelaars A, van Bel F, Heijnen CJ. Mesenchy-
mal stem cell transplantation changes the gene expression profile
37. Daadi MM, Davis AS, Arac A, et al. Human neural stem cell grafts
of the neonatal ischemic brain. Brain Behav Immun 2011;25:
modify microglial response and enhance axonal sprouting in neo-
1342–1348.
natal hypoxic-ischemic brain injury. Stroke 2010;41:516–523.
55. Bliss TM, Andres RH, Steinberg GK. Optimizing the success of cell
38. Ma J, Wang Y, Yang J, et al. Treatment of hypoxic-ischemic ence-
transplantation therapy for stroke. Neurobiol Dis 2010;37:
phalopathy in mouse by transplantation of embryonic stem cell-
275–283.
derived cells. Neurochem Int 2007;51:57–65.
56. Kassis I, Grigoriadis N, Gowda-Kurkalli B, et al. Neuroprotection
39. Park KI, Teng YD, Snyder EY. The injured brain interacts recipro-
and immunomodulation with mesenchymal stem cells in chronic
cally with neural stem cells supported by scaffolds to reconstitute
experimental autoimmune encephalomyelitis. Arch Neurol 2008;
lost tissue. Nat Biotechnol 2002;20:1111–1117.
65:753–761.
40. Yasuhara T, Matsukawa N, Yu G, et al. Behavioral and histological
57. Fainstein N, Vaknin I, Einstein O, et al. Neural precursor cells in-
characterization of intrahippocampal grafts of human bone mar-
hibit multiple inflammatory signals. Mol Cell Neurosci 2008;39:
row-derived multipotent progenitor cells in neonatal rats with
335–341.
hypoxic-ischemic injury. Cell Transplant 2006;15:231–238.
58. Ben-Hur T. Immunomodulation by neural stem cells. J Neurol Sci
41. Yasuhara T, Hara K, Maki M, et al. Intravenous grafts recapitulate
2008;265:102–104.
the neurorestoration afforded by intracerebrally delivered multipo-
tent adult progenitor cells in neonatal hypoxic-ischemic rats. J 59. Einstein O, Fainstein N, Vaknin I, et al. Neural precursors attenu-
Cereb Blood Flow Metab 2008;28:1804–1810. ate autoimmune encephalomyelitis by peripheral immunosuppres-
sion. Ann Neurol 2007;61:209–218.
42. van Velthoven CT, Kavelaars A, van Bel F, Heijnen CJ. Mesenchy-
mal stem cell treatment after neonatal hypoxic-ischemic brain 60. Borlongan CV, Hadman M, Sanberg CD, Sanberg PR. Central
injury improves behavioral outcome and induces neuronal and oli- nervous system entry of peripherally injected umbilical cord blood
godendrocyte regeneration. Brain Behav Immun 2010;24: cells is not required for neuroprotection in stroke. Stroke 2004;35:
387–393. 2385–2389.

May 2012 599


ANNALS of Neurology

61. Willing AE, Lixian J, Milliken M, et al. Intravenous versus intrastria- 70. Miller SP, Ferriero DM, Leonard C, et al. Early brain injury in pre-
tal cord blood administration in a rodent model of stroke. J Neu- mature newborns detected with magnetic resonance imaging is
rosci Res 2003;73:296–307. associated with adverse early neurodevelopmental outcome. J
Pediatr 2005;147:609–616.
62. Zawadzka M, Lukasiuk K, Machaj EK, et al. Lack of migration and
neurological benefits after infusion of umbilical cord blood cells in 71. Back SA, Luo NL, Mallinson RA, et al. Selective vulnerability of
ischemic brain injury. Acta Neurobiol Exp (Wars) 2009;69:46–51. preterm white matter to oxidative damage defined by F2-isopros-
tanes. Ann Neurol 2005;58:108–120.
63. Nystedt J, Makinen S, Laine J, Jolkkonen J. Human cord blood
CD34þ cells and behavioral recovery following focal cerebral is- 72. Noble M, Davies JE, Mayer-Proschel M, et al. Precursor cell biology
chemia in rats. Acta Neurobiol Exp (Warsz) 2006;66:293–300. and the development of astrocyte transplantation therapies: lessons
from spinal cord injury. Neurotherapeutics 2011;8:677–693.
64. Makinen S, Kekarainen T, Nystedt J, et al. Human umbilical cord
blood cells do not improve sensorimotor or cognitive outcome 73. Volpe JJ. Brain injury in premature infants: a complex amalgam of
following transient middle cerebral artery occlusion in rats. Brain destructive and developmental disturbances. Lancet Neurol 2009;
Res 2006;1123:207–215. 8:110–124.
65. Moodley Y, Ilancheran S, Samuel C, et al. Human amnion epithe- 74. Buser JR, Maire J, Riddle A, et al. Arrested pre-oligodendrocyte
lial cell transplantation abrogates lung fibrosis and augments maturation contributes to myelination failure in premature infants.
repair. Am J Respir Crit Care Med 2010;182:643–651. Ann Neurol. DOI: 10.1002/ana.22627, 2011.
66. Grigsby PL, Hirst JJ, Scheerlinck JP, et al. Fetal responses to 75. Yager JY, Ashwal S. Animal models of perinatal hypoxic-ischemic
maternal and intra-amniotic lipopolysaccharide administration in brain damage. Pediatr Neurol 2009;40:156–167.
sheep. Biol Reprod 2003;68:1695–1702.
76. Park KI, Himes BT, Stieg PE, et al. Neural stem cells may be uniquely
67. Nitsos I, Rees SM, Duncan J, et al. Chronic exposure to intra-am- suited for combined gene therapy and cell replacement: evidence
niotic lipopolysaccharide affects the ovine fetal brain. J Soc Gyne- from engraftment of neurotrophin-3-expressing stem cells in
col Investig 2006;13:239–247. hypoxic-ischemic brain injury. Exp Neurol 2006;199:179–190.
68. Lu P, Jones LL, Snyder EY, Tuszynski MH. Neural stem cells consti- 77. Qu SQ, Luan Z, Yin GC, et al. Transplantation of human fetal neu-
tutively secrete neurotrophic factors and promote extensive host ral stem cells into cerebral ventricle of the neonatal rat following
axonal growth after spinal cord injury. Exp Neurol 2003;181: hypoxic-ischemic injury: survival, migration and differentiation [in
115–129. Chinese]. Zhonghua Er Ke Za Zhi 2005;43:576–579.
69. Committee on Understanding Premature Birth and Assuring 78. Xia G, Hong X, Chen X, et al. Intracerebral transplantation of mes-
Healthy Outcomes. In: Behrman RE, Butler AS, eds. Preterm birth: enchymal stem cells derived from human umbilical cord blood
causes, consequences, and prevention. Washington DC: Institute alleviates hypoxic ischemic brain injury in rat neonates. J Perinat
of Medicine of the National Academies, 2007, pp. 346–397. Med 2010;38:215–221.

600 Volume 71, No. 5

You might also like