ArticlePDF Available

Spastic Paresis After Perinatal Brain Damage in Rats Is Reduced by Human Cord Blood Mononuclear Cells

Authors:

Abstract and Figures

Brain damage around birth may cause lifelong neurodevelopmental deficits. We examined the therapeutic potential of human umbilical cord blood-derived mononuclear cells containing multipotent stem cells to facilitate motor recovery after cerebral hypoxic-ischemic damage in neonatal rats. Left carotid artery ligation followed by 8% O(2) inhalation for 80 min was performed on postnatal d 7, succeeded by intraperitoneal transplantation of human umbilical cord blood-derived mononuclear cells on postnatal d 8 in a sham-controlled design. Histologic and immunohistochemical analysis on postnatal d 21 revealed that neonates developed severe cerebral damage after the hypoxic-ischemic insult. These animals also suffered from contralateral spastic paresis, as evidenced by their locomotor behavior. After transplantation of human umbilical cord blood-derived mononuclear cells, spastic paresis was largely alleviated, resulting in a normal walking behavior. This "therapeutic" effect was accompanied by the fact that mononuclear cells had entered the brain and were incorporated around the lesion without obvious signs of transdifferentiation. This study demonstrates that intraperitoneal transplantation of human umbilical cord blood-derived mononuclear cells in a rat model of perinatal brain damage leads to both incorporation of these cells in the lesioned brain area and to an alleviation of the neurologic effects of cerebral palsy as assessed by footprint and walking pattern analysis.
Content may be subject to copyright.
Spastic Paresis After Perinatal Brain Damage in Rats Is Reduced
by Human Cord Blood Mononuclear Cells
CAROLA MEIER, JOHANNES MIDDELANIS, BIANCA WASIELEWSKI, SANDRA NEUHOFF, ASTRID ROTH-HAERER,
MARKUS GANTERT, HUBERT R. DINSE, ROLF DERMIETZEL, AND ARNE JENSEN
Department of Neuroanatomy and Molecular Brain Research [C.M., B.W., S.N., A.R.-H., R.D.], Department of Neuroinformatics [H.R.D.],
Ruhr-University Bochum, D- 44801 Bochum, Germany; Department of Gynecology and Obstetrics [J.M., M.G., A.J.], Ruhr-University
Bochum, D- 44892 Bochum, Germany
ABSTRACT: Brain damage around birth may cause lifelong neu-
rodevelopmental deficits. We examined the therapeutic potential of
human umbilical cord blood– derived mononuclear cells containing
multipotent stem cells to facilitate motor recovery after cerebral
hypoxic-ischemic damage in neonatal rats. Left carotid artery ligation
followed by 8% O
2
inhalation for 80 min was performed on postnatal
d 7, succeeded by intraperitoneal transplantation of human umbilical
cord blood– derived mononuclear cells on postnatal d 8 in a sham-
controlled design. Histologic and immunohistochemical analysis on
postnatal d 21 revealed that neonates developed severe cerebral
damage after the hypoxic-ischemic insult. These animals also suf-
fered from contralateral spastic paresis, as evidenced by their loco-
motor behavior. After transplantation of human umbilical cord
blood– derived mononuclear cells, spastic paresis was largely allevi-
ated, resulting in a normal walking behavior. This “therapeutic”
effect was accompanied by the fact that mononuclear cells had
entered the brain and were incorporated around the lesion without
obvious signs of transdifferentiation. This study demonstrates that
intraperitoneal transplantation of human umbilical cord blood–
derived mononuclear cells in a rat model of perinatal brain damage
leads to both incorporation of these cells in the lesioned brain area
and to an alleviation of the neurologic effects of cerebral palsy as
assessed by footprint and walking pattern analysis. (Pediatr Res 59:
244–249, 2006)
Each year, thousands of children incur perinatal brain
damage that potentially results in lifelong sequelae (1,2).
Depending on the extent and location of the insult these
children may develop spastic paresis, choreo-athetosis, ataxia,
and disorders of sensorimotor coordination (1). In the United
States, estimates of the costs to society for treatment and care
of these children amount to a total of $11.5 billion per birth
year (2).
One of the major causes of perinatal brain damage is severe
fetal or neonatal asphyxia (3,4), resulting in cerebral hypoxic-
ischemic insults and hemorrhages. Experimental evidence has
shown that neuroprotective strategies using pharmacologic
agents or moderate cerebral hypothermia may ameliorate peri-
natal brain damage (5,6). However, limited clinical strategies
are available for functional regeneration of damaged nervous
structures in the perinatal period (7). Therefore, one of the
most urgent tasks for scientists and clinicians will be to
explore the enormous potential of cell therapies using stem
cells in general and hUCB-derived cells in particular to pro-
vide a therapeutic paradigm for perinatal neuronal repair.
For hUCB cell transplantation, a number of promising
experimental protocols have been established in sheep (8) and
rats (9). The study by Chen et al. (9) provides first experi-
mental evidence in adult rats that intravenous application of
umbilical cord blood cells results in the migration of these
cells toward brain regions affected by stroke. Furthermore,
behavioral studies on those animals revealed an at least partial
functional compensation.
The insult resulting from perinatal hypoxic-ischemic brain
damage in humans has been reproduced in a neonatal rat
model (Levine model) (10 –12). Here, we present a study
designed to investigate the potential of hUCB-derived mono-
nuclear cells to migrate to a hypoxic-ischemic brain lesion
from a distant transplantation site, and to examine their po-
tential to alleviate neurologic deficits in neonatal rats. Our
results demonstrate that intraperitoneal transplantation of
hUCB-mononuclear cells after a hypoxic-ischemic insult re-
sults both in the migration of these cells toward the lesion in
the neonatal rat brain and in the recovery from spastic paresis
as assessed by walking pattern analysis.
MATERIALS AND METHODS
hUCB-derived mononuclear cells. Blood from umbilical cord and pla-
centa was obtained from the Department of Gynecology and Obstetrics
(Ruhr-University Bochum, Germany), after receiving the mother’s informed
consent. The umbilical vein was punctured post partum, and the blood was
collected in umbilical cord blood collection bags containing citrate phosphate
dextrose as an anticoagulant (Maco Pharma, Langen, Germany) and stored at
room temperature up to 24 h until further processing.
Preparation of the mononuclear cell fraction was performed by Ficoll
Paque (Amersham, Freiburg, Germany) density gradient centrifugation ac-
Received March 17, 2005; accepted September 21, 2005.
Correspondence: Arne Jensen, M.D., Department of Gynecology and Obstetrics,
Ruhr-University Bochum, In der Schornau 23-25, D-44892 Bochum, Germany; e-mail:
Arne.Jensen@rub.de
Supported by grants of the Stem Cell Network North Rhine Westphalia (C.M., A.J.)
and by the Medical Faculty of Ruhr-University Bochum (FoRUM) (A.J.).
DOI: 10.1203/01.pdr.0000197309.08852.f5
Abbreviations: GFAP, glial fibrillary acidic protein; HLA-DR, human leu-
kocyte antigen type DR; hUCB, human umbilical cord blood
0031-3998/06/5902-0244
PEDIATRIC RESEARCH Vol. 59, No. 2, 2006
Copyright © 2006 International Pediatric Research Foundation, Inc. Printed in U.S.A.
244
cording to the manufacturer’s instructions. The mononuclear fraction of cells
was collected from the interphase, resuspended in 0.9% sodium chloride, and
the cell number was determined. Viability of resuspended mononuclear cells
was 97 62%.
Cerebral ischemia. The Levine model (10,13) was used to achieve repro-
ducible hypoxic-ischemic injury in neonatal rats. Seven-day-old Wistar rat
pups were deeply anesthetized by inhalation of 4% halothane and maintained
with 1.5% halothane in 50% N
2
O/50% O
2
. The left common carotid artery
was exposed, double-ligated with 6 0 surgical silk, and severed. The duration
of anesthesia and surgery did not exceed 10 min. After surgery, the rat pups
were allowed to recover in their home cages for 1 h. To introduce systemic
hypoxia, the pups were subsequently placed in an incubator (Incubator 7510;
Dra¨ger, Lu¨beck, Germany) and exposed to a hypoxic gas mixture (8%
oxygen, 92% nitrogen) for 80 min. The environmental temperature was
strictly maintained at 36°C.
All surgical and experimental protocols were approved by the appropriate
institutional review committee (Bezirksregierung Arnsberg, Germany) and
met the guidelines of the German animal protection law.
Experimental protocol. Randomly selected animals were assigned to three
different experimental groups: controls (no lesion), lesion (without transplan-
tation), and lesion followed by transplantation of mononuclear cells.
Animals of the lesion group (lesion only, n511) were subjected to
ligation of the left common carotid artery, followed by systemic hypoxia.
Twenty-four hours after the insult, this group received an intraperitoneal
injection of 500
m
L of 0.9% sodium chloride (sham injection). In the
transplantation group (lesion followed by transplantation, n514), rats
received 1 310
7
hUCB-derived mononuclear cells (in a volume of 500
m
L
0.9% sodium chloride) by intraperitoneal injection 24 h after the hypoxic-
ischemic insult. The control group (no lesion, n511) comprised of animals
that did not undergo any treatment or surgery (n55), and of sham-operated
animals that were anesthetized, the carotid artery exposed but not ligated, and
surgery then terminated (n56). After initial analysis, these latter two groups
were indistinguishable in motor analysis, and therefore combined. Additional
control animals (no lesion, n510), which received intraperitoneal injection
of 1 310
7
hUCB-derived mononuclear cells, were included in motor
analysis. There was no use of immunosuppressants or analgesics in any
experimental group. Animals were included into subsequent analyses when
displaying a severe macroscopic lesion accompanied by cystic changes larger
than 4 mm upon hypoxic-ischemic damage without (11 out of 18 animals) or
with (14 out of 20 animals) transplantation of hUCB-derived cells, thereby
disregarding animals in which the Levine procedure resulted in minor lesions.
For the control group, animals were included when displaying an intact brain
without pigmentation or cystic changes (11 out of 13 animals).
Analysis of motor abilities. Footprint analysis was performed on postnatal
d 21 in a strictly blinded fashion. The paws were colored consecutively, and
the footprints were printed onto paper when the rat walked along a defined
gangway which was elevated by 35°. Footprints were then measured and each
imprint was analyzed regarding the distance between the first and fifth toe
(hind paws) or between the first and the fourth digit (forepaws) (14). In
addition, the step length was determined for each paw. A significant decrease
in footprint width and/or step length of the limbs contralateral to the lesion
compared with that of the ipsilateral site was considered to reflect spastic
paresis.
Macroscopic assessment. Rats were anesthetized by CO
2
exposition and
decapitated at postnatal d 21. Brains were dissected and brain injury was
assessed immediately after dissecting the brains (11). Neonatal rats were
considered to have suffered hypoxic-ischemic brain damage when displaying
a cystic infarction, similar to a unilateral internal hydrocephalus, on the brain
surface ipsilateral to the carotid artery occlusion.
Microscopic assessment. Brains were covered in tissue freezing medium
(Leica, Nussloch, Germany) and frozen in 8% methylcyclohexane in 2-methyl-
butan (vol/vol) (– 80°C). Histology and immunohistochemistry were per-
formed on cryosections of 14-
m
m thickness, which had been mounted onto
Superfrost Plus slides (BDH, London, UK) and heat dried for 1 h at 40°C
before fixation.
Precise localization of the lesion areas was assured by histologic staining
according to Klu¨ver-Barrera, showing myelinated fiber tracts in light green
and cell bodies in blue. The lesion size was determined by measuring the
hemisphere areas in histologic sections. Measurements were performed in
nine histologic sections at three defined levels in the brain, and in three
animals per group (control, lesion, lesion plus transplantation). The extent of
brain damage was expressed as area of the left (lesioned) hemisphere as
percentage of the area of the right (nonlesioned) hemisphere.
For immunohistochemistry, adjacent cryosections were fixed in –20°C
cold ethanol, rinsed in PBS and preincubated in blocking buffer (BB, 10%
normal goat serum and 0.1% TritonX-100 in PBS) for 30 min. Primary
antibodies were diluted in BB and sections were incubated at 18°C overnight.
Samples were rinsed in PBS and incubated in 0.2% BSA in PBS. Incubation
with secondary antibodies was performed in BB at 18°C for 2 h. Sections
were mounted using the ProLong Antifade Kit (Molecular Probes, Leiden,
Netherlands). Fluorescence was documented using confocal imaging micros-
copy (Zeiss LSM 510 META). Data were collected using the single track
scanning module and exported as TIFF files into Adobe Photoshop 7.0
(Adobe Imaging Systems Inc., Mountain View, CA).
Primary antibodies (dilution and supplier in parentheses) were directed
against HLA-DR
a
-chain (1:50; DAKO, Hamburg, Germany), cleaved
caspase3 (1:100; New England Biolabs, Frankfurt, Germany), CD68 (ED1;
1:100; Serotec, Du¨sseldorf, Germany), glial fibrillary acidic protein (GFAP;
monoclonal: 1:100, Chemicon, Hofheim, Germany; polyclonal: 1:100, Sigma
Chemical Co., Taufkirchen, Germany). Primary antibodies used to determine
neuronal differentiation of HLA-DR-labeled hUCB-derived mononuclear
cells included the polyclonal GFAP antibody, neurofilament-68 (Chemicon),
and synaptophysin antibodies (Diagnostic Biosystems, Pleasanton, CA). Re-
activity of these antibodies with human cells had been confirmed in vitro (data
not shown).
Secondary antibodies were Alexa Fluor 488 or 546 conjugated goat-anti-
rabbit or goat-anti-mouse (1:3,000; Molecular Probes).
Statistical analysis. The Kolmogorov-Smirnov test was performed to
demonstrate that test distributions were normal. Further analysis of intra- and
intersample variability was done by two-way ANOVA (SuperANOVA; Aba-
cus Concepts Inc., Berkeley, CA), followed by the appropriate posthoc test
(Student-Newman-Keuls). Data are expressed as mean 6SEM. A value of p
,0.05 was considered statistically significant.
RESULTS
Hypoxic-ischemic brain damage in perinatal rats. Cere-
bral hypoxia-ischemia resulted in macroscopic changes, in-
cluding cystic lesions (n511). There was no evidence of any
brain damage in control rats (n512). The rostrocaudal extent
of the damage was determined and averaged 6.1 60.6 mm in
lesioned brains. Histopathological analysis revealed that hy-
poxic-ischemic brain damage affected cortex, hippocampus,
and periventricular areas (Fig. 1, A–C), and comprised inflam-
mation and neuronal cell death (see below).
The extent of inflammation was determined by CD68-
immunopositive activated microglia (15). Apoptotic neuronal
cell death was detected by cleaved-caspase3 immunoreactivity
(16). There were activated microglia (green) in a large region
of the lesioned hemisphere, whereas the apoptotic neuronal
cells were clustered within (red), as depicted in Figure 2, A
and B. Intact areas of the ipsilateral hemisphere as well as the
contralateral hemisphere were devoid of any immunoreactiv-
ity to CD68, indicating the absence of inflammation (Fig. 2, A
and C; green). However, weak cleaved-caspase3 immunore-
activity was detectable in intact hemispheres (Fig. 2, Aand C;
red), as described previously during brain development (17).
“Homing” of human umbilical mononuclear cells. Trans-
plantation of human mononuclear cells (1 310
7
) derived
Figure 1. Histology of the hypoxic-ischemic brain lesion. (A) The level of
coronal sections shown in Band Cis indicated in a schematic drawing. (B)
Histologic staining (Klu¨ver-Barrera) reveals damage of the left brain hemi-
sphere, i.e. ipsilateral to the carotid artery occlusion. The lesion affects the
hippocampus (hc), parietal and temporal cortex (cx), periventricular areas
(pv), and results in an enlarged lateral ventricle (v). (C) Schematic represen-
tation of the histologic features observed in B. Boxed areas were analyzed
immunohistochemically, shown in Figures 2 and 3. Scale bar: 1 mm.
245UMBILICAL CELLS REDUCE SPASTIC PARESIS
from umbilical cord blood was performed by intraperitoneal
injection 24 h after the hypoxic-ischemic insult, i.e. in post-
natal d 8 rats (n514), in a sham-controlled fashion. Cells
were identified by immunohistochemical detection of human-
specific HLA-DR
a
-chain surface antigens (18). Thus, we
demonstrate that transplanted human mononuclear cells mi-
grate from the intraperitoneal cavity to the damaged brain
region (Fig. 3, Aand B; green). These cells were detectable in
the brain as early as 3 d after intraperitoneal transplantation
and were still present in the lesioned hemisphere 2 wk after
transplantation (Fig. 3, Aand B; green). Confocal microscopy
revealed that transplanted cells (green immunofluorescence in
Fig. 3B) were clearly incorporated into the astrocytic network
(red immunofluorescence in Fig. 3, B–D). In contrast, there
was no evidence for human cells outside the damaged brain
region (Fig. 3C), and there was no HLA immunoreactivity in
the lesioned areas of animals that had not received transplan-
tation of hUCB-derived mononuclear cells (Fig. 3D). Interest-
ingly, transplanted human cells found in the brain were strictly
confined to the area of activated microglia (compare Fig. 2A;
green).
Transplantation of hUCB cells did not change the severity
of morphologic damage. In control animals, left and right
hemisphere comprised approximately the same area. Upon
hypoxic-ischemic brain injury, the area of the lesioned hemi-
sphere was 34.99 64.7% of that of the intact hemisphere. In
lesioned animals that had received transplantation of hUCB-
derived mononuclear cells, the lesioned hemisphere area com-
prised 35.72 60.4% of the intact hemisphere area, which was
not significantly different from nontransplanted animals (p$
0.05). In addition, HLA-DR-immunopositive cells did not
display obvious signs of transdifferentiation in vivo, as as-
sessed by their phenotype and the absence of GFAP, an
intermediate filament expressed in neural progenitor cells, and
neuron-specific proteins like neurofilament-68 and synapto-
physin (Fig. 4), although expression of these markers was
observed upon cultivation of hUCB-derived mononuclear
cells in the presence of nerve growth factor and retinoic acid
in vitro (data not shown).
Perinatal brain damage causes spastic paresis. To exam-
ine whether transplantation of hUCB-derived mononuclear
cells after cerebral hypoxic-ischemic brain damage has bene-
ficial effects on locomotor behavior, we performed a strictly
blinded footprint and walking pattern analysis. Our analysis of
the toe distance 1 to 5 of the hind paw contralateral to the
cerebral damage at 3 wk of age revealed a highly significant
reduction (p,0.001) of this measure after cerebral damage
(0.99 60.04 cm; n510) compared with controls (1.15 6
0.04 cm; n512), which is characteristic for spastic paresis.
Figure 2. Activated microglia and apoptotic neuronal cells are located in the
vicinity of the hypoxic-ischemic lesion. (A) Schematic representation of the
lesion site as indicated by the presence of CD68-immunopositive cells (green)
and cleaved-caspase3 expressing cells (red). Immunohistochemistry is pre-
sented for areas Band C. (B) Immunohistochemistry for CD68 (green
fluorescence), showing activated microglia, and cleaved-caspase3 immunore-
activity (red fluorescence) indicating apoptosis in the lesioned hemisphere.
(C) The nonlesioned hemisphere is devoid of activated microglia (green
fluorescence) and shows few weakly labeled caspase3-positive cells as char-
acteristic for neonatal rat brain. Scale bar: 100
m
m.
Figure 3. Intraperitoneal transplantation of mononuclear hUCB-derived
cells resulted in a specific “homing” of these cells into the CNS and
incorporation around the lesioned area. (A) Schematic representation of the
distribution of human cells in the lesioned hemisphere. HLA-DR-positive
cells (green) are detected in the area of the hypoxic-ischemic lesion. Con-
tralateral hemispheres were devoid of human cells. Immunohistochemistry is
demonstrated for boxed areas in B(lesioned hemisphere) and C(nonlesioned
hemisphere). (B) HLA-DR-positive mononuclear cells (green) are located
within a scaffold of GFAP-positive astrocytes (red) in the area of the
hypoxic-ischemic lesion. (C) Nonlesioned parts of the brain, as demonstrated
by the absence of inflammatory and apoptotic events (compare Fig. 2), are
devoid of human cells (absence of green fluorescence). (D) Lesioned areas of
the brain of animals that did not receive transplantation of human cells are
devoid of HLA-immunostaining. Note absence of green immunofluorescence
(HLA-staining) within the red scaffold of GFAP-positive rat astrocytes. Scale
bar in B, C: 100
m
m; in D: 20
m
m.
246 MEIER ET AL.
As animals after hypoxic-ischemic lesion were smaller (43.2
63.2 g) than control animals (50.3 63.1 g) of the same age,
an intraindividual comparison of footprint width was per-
formed. In control animals (without transplantation), there
was no significant difference in toe distance between left (1.16
60.03 cm) and right (1.15 60.04 cm; p$0.05; Fig. 5A) hind
paws. In nonlesioned control animals, transplantation of
hUCB cells did not result in significant differences in the toe
distance of left (1.19 60.01 cm) and right (1.21 60.01 cm;
p$0.05) hind paws. However, in lesioned animals (without
transplantation), the toe distance of the ipsilateral hind paw
(1.12 60.03 cm) differed significantly from that of the
contralateral hind paw (0.99 60.04 cm; p,0.05; Fig. 5A),
and this difference is independent of the animal’s weight.
Furthermore, in animals with hypoxic-ischemic lesion,
there was evidence for a reduction of the step length of the
right hind paw (contralateral to the lesion; 7.57 60.07 cm)
compared with the left hind paw controls (8.26 60.33 cm; p
,0.05; Fig. 5B), which is also characteristic of spastic paresis
(19).
Intraperitoneal transplantation of umbilical mononuclear
cells reduces spastic paresis. Most importantly, our results
show that these neurologic deficits were eliminated following
transplantation of hUCB mononuclear cells. There was a
Figure 4. Transplanted HLA-immunopositive human UCB-derived mononu-
clear cells do not reveal signs of transdifferentiation upon “homing” to the
hypoxic-ischemic lesion. (A, B) GFAP (red), characteristic for astrocytes as
well as neuroglial progenitor cells, is located in close vicinity to human
HLA-DR-positive cells (green), however, there is no obvious overlap be-
tween GFAP- and HLA-DR-immunofluorescence. The neuron-specific pro-
teins NF-68 (C, D) and synaptophysin (E, F) are expressed in the brain tissue
(red fluorescence), however, HLA-positive cells (green fluorescence) are
devoid of NF-68 and synaptophysin immunostaining. Scale bar in A, C, E: 20
m
m; B, D, F: 50
m
m.
Figure 5. Transplantation of hUCB-derived mononuclear cells reduces spas-
tic paresis as assessed by footprint analysis of 3-wk-old animals. (A) Hypoxic-
ischemic brain damage results in spastic paresis of the distal limb muscles,
causing a significant reduction of footprint width (toe distance 1 to 5) of the
right hind paw (contralateral to the insult; black columns) compared with the
left (ipsilateral; gray columns) hind paw. Intraperitoneal transplantation of
hUCB-derived mononuclear cells after hypoxic-ischemic brain damage re-
duced spastic paresis. In these animals, differences between ipsi- and con-
tralateral hind paws were no longer detectable. Photographs of footprints
(right hind paws) illustrate the footprint widths (arrows) of control animals
without (left) and with (center left) transplantation, upon hypoxic-ischemic
lesion without (center right) and with (right) transplantation of hUCB-derived
mononuclear cells. (B) In control animals with and without transplantation,
the step length of left and right hind paws is equal. In contrast, hypoxic-
ischemic lesion resulted in a significantly reduced step length of the right hind
paw (black columns) compared with the left hind paw (gray columns). This
reduction in step length of the hind paw contralateral to the lesion, also
indicative of spastic paresis, was largely alleviated upon transplantation of
hUCB-derived mononuclear cells. Data are presented as mean 6SEM; *p,
0.05; ***p,0.001.
247UMBILICAL CELLS REDUCE SPASTIC PARESIS
dramatic alleviation of the contralateral spastic paresis for all
parameters back to normal (Fig. 5). Upon transplantation,
there was no significant difference detected between toe dis-
tances 1–5 of the ipsilateral (1.20 60.03 cm) and contralateral
(1.21 60.04 cm) hind paws of animals after hypoxic-
ischemic lesion and transplantation (p$0.05). The step
length of the ipsilateral (8.46 60.4 cm) hind paws also
equaled that measured on the contralateral side (8.40 60.32
cm; p$0.05). Interestingly, the weight of these animals (51.4
61.4 g) was similar to that of control animals, however, it
differed significantly from that of lesioned animals that had
not received hUCB-cell transplantation (p,0.05).
DISCUSSION
In this study, we used a model of perinatal hypoxic-
ischemic brain damage in rats. The insult was morphologically
characterized by hemorrhage, neuronal death, and inflamma-
tion, and functionally by spastic paresis of contralateral fore-
and hind limbs. Intraperitoneal transplantation of human
UCB-derived mononuclear cells resulted in a) migration of
cells from the peritoneal cavity to the CNS, b) incorporation of
cells around the cerebral lesion (“homing”), and c) an allevi-
ation of spastic paresis.
Unlike other studies, we used the intraperitoneal cavity for
transplantation. The migration of transplanted cells from the
intraperitoneal cavity to the damaged region of the brain
suggests the presence of very specific and powerful chemoat-
tractant signals. One component of the complex mechanism
seems to be the expression of cytokines/chemokines in the
brain in response to hypoxic-ischemic brain injury (20). The
chemoattractant effect of adult ischemic brain tissue had pre-
viously been demonstrated by cell migration assays, in which
hUCB-derived cells were shown to migrate towards tissue
extract derived from lesioned brain hemispheres (9). In our
paradigm, invasion of mononuclear cells was very focused in
that it was confined to the hypoxic-ischemic region of the
brain. More general patterns of brain damage as observed in
human genetic metabolic disorders like Krabbe leukodystro-
phy lead to a more widespread distribution of damaged neu-
rons, and hence result in a ubiquitous distribution of trans-
planted human hUCB cells (21,22).
The specificity of “homing” within the hypoxic-ischemic
brain may both be linked to specific chemoattractants (20) and
to the lack of a functioning blood-brain-barrier in the damaged
brain regions (13,23) leading to a facilitated intrusion of
human mononuclear cells into damaged regions and to a
preferential regional incorporation of these cells. Furthermore,
successful “homing” might be related to the early transplan-
tation after the insult, because then concentrations of chemoat-
tractants are likely to be maximal, and, secondly, there is a
known optimal point in time for transplantation in rodents
within the first 2 wk after the insult (24).
Successful “homing” may be also related to the use of
crude, rather than predifferentiated, hUCB-derived mononu-
clear cells for transplantation in this study, resulting in a
minimal manipulation of cells, which was limited to density
gradient centrifugation and resuspension in sodium chloride
only. Thus, the full potential of cell (sub-) populations and that
of cellular responsiveness to sense signals important to entrain
migration to the damaged brain region may have been pre-
served.
Transplantation of human mononuclear cells into neonatal
rats also warrants some considerations concerning graft– host
interactions. Our results suggest some degree of immune
tolerance toward cord blood cells, possibly mediated by di-
minished generation of cytotoxic responses of the host (25)
through IL-10 release by these cells (26).
Neurologic deficits like contralateral spastic paresis and
reduced step length are characteristic of pyramidal tract im-
pairment in human newborns. Thus, the most important issue
in any therapeutic approach, i.e. functional recovery, was
achieved in our model. Footprint analysis represents one of the
most sensitive parameters to detect pyramidal motor dysfunc-
tion in early neonatal life (19).
Irrespective of the striking “therapeutic” effect of mononu-
clear cell transplantation that also included the normalization
of the weight, the underlying mechanisms remain largely
unknown. Interestingly, we produced no evidence for neural
differentiation of these transplanted human cells in the rat
brain, even though hUCB mononuclear cells in our hands
showed expression of a variety of neural markers in vitro
using differentiation media (data not shown). This appears in
part at variance with observations by Zigova et al. (27).
However, this study is not directly comparable due to the
methods used by those authors, i.e. predifferentiation of hUCB
cells, intracisternal injection of cells, and the use of P1 rats. In
our study, there was no change in the phenotype of HLA-
positive mononuclear cells or in the expression of glial or
neuronal marker proteins even though there was clear struc-
tural integration of the human cells into the three-dimensional
astrocytic scaffold of the rat brain. None of the neural markers
used yielded positive immunohistochemical staining of HLA-
positive transplanted human cells. Although we were able to
demonstrate a strong therapeutic effect of the transplantation
by largely alleviating the neurologic deficits observed in rats
after cerebral hypoxic ischemia without transplantation, the
beneficial functional outcome appears to be unrelated to true
cell replacement by differentiation of transplanted human
cells. Thus, a permissive effect is suggested, as observed in the
lesioned heart (28) and brain (29). Hence, in our model of
hypoxic-ischemic brain damage, we propose secondary mech-
anisms, which may reduce adverse effects resulting from
neural damage per se. Tentative candidates for these second-
ary mechanisms are increased vascularization, reduced edema,
detoxification, prevention of gliosis, and others. Also, “by-
stander effects” mediated by other trophic factors, including
glial cell line– derived neurotrophic factor, brain-derived neu-
rotrophic factor, and nerve growth factor, may contribute to
functional recovery caused e.g. by extensive host axonal
growth and increased neuronal survival (30). Furthermore,
recruitment of endogenous neural stem cells, e.g. elicited
through cytokine and growth factor release, ought to be
considered.
In summary, we show in a neonatal cerebral hypoxia-
ischemia model that hUCB-derived mononuclear cells trans-
248 MEIER ET AL.
planted intraperitoneally enter the rat brain and incorporate
specifically around the lesion (“homing”) in large numbers.
Transplantation results in substantial alleviation of spastic
paresis as assessed by footprint recordings of the hind paw
contralateral to the damaged brain hemisphere and walking
track analysis. This study provides important information for
the development of therapeutic strategies using hUCB stem
cells after perinatal brain damage to reduce potential sensori-
motor deficits.
Acknowledgments. The authors thank Nicole-Christiane
Kozik, Hans-Werner Habbes, Janet Moers, and Kerstin
Schmitz for excellent technical assistance, and Petra Paraken-
ings and Helga Schulze for expertly done photographic work.
REFERENCES
1. Volpe JJ 2001 Neurology of the Newborn. WB Saunders, Philadelphia, pp 217–396
2. Centers for Disease Control and Prevention (CDC) 2003 Economic Costs Associated
with Mental Retardation, Cerebral Palsy, Hearing Loss, and Vision Impairment—
United States. MMWR Morb Mortal Wkly Rep 53:57–59
3. Jensen A, Berger R 1991 Fetal circulatory responses to oxygen lack. J Dev Physiol
16:181–207
4. Jensen A, Garnier Y, Middelanis J, Berger R 2003 Perinatal brain damage—from
pathophysiology to prevention. Eur J Obstet Gynecol Reprod Biol 110:S70–S79
5. Hamrick SE, Ferriero DM 2003 The injury response in the term newborn brain: can
we neuroprotect? Curr Opin Neurol 16:147–154
6. Vannucci RC, Connor JR, Mauger DT, Palmer C, Smith MB, Towfighi J, Vannucci
SJ 1999 Rat model of perinatal hypoxic-ischemic brain damage. J Neurosci Res
55:158–163
7. Gluckman PD, Wyatt JS, Azzopardi D, Ballard R, Edwards AD, Ferriero DM, Polin
RA, Robertson CM, Thoresen M, Whitelaw A, Gunn AJ 2005 Selective head cooling
with mild systemic hypothermia after neonatal encephalopathy: multicentre random-
ised trial. Lancet 365:663–670
8. Kogler G, Sensken S, Airey JA, Trapp T, Muschen M, Feldhahn N, Liedtke S, Sorg
RV, Fischer J, Rosenbaum C, Greschat S, Knipper A, Bender J, Degistirici O, Gao
J, Caplan AI, Colletti EJ, Almeida-Porada G, Muller HW, Zanjani E, Wernet P 2004
A new human somatic stem cell from placental cord blood with intrinsic pluripotent
differentiation potential. J Exp Med 200:123–135
9. Chen J, Sanberg PR, Li Y, Wang L, Lu M, Willing AE, Sanchez-Ramos J, Chopp
M 2001 Intravenous administration of human umbilical cord blood reduces behav-
ioral deficits after stroke in rats. Stroke 32:2682–2688
10. Levine S 1960 Anoxic-ischemic encephalopathy in rats. Am J Pathol 36:1–17
11. Bona E, Johansson BB, Hagberg H 1997 Sensorimotor function and neuropathology
five to six weeks after hypoxia-ischemia in seven-day-old rats. Pediatr Res 42:678–
683
12. Hagberg H, Gilland E, Diemer NH, Andine P 1994 Hypoxia-ischemia in the neonatal
rat brain: histopathology after post-treatment with NMDA and non-NMDA receptor
antagonists. Biol Neonate 66:205–213
13. Rice JE 3rd, Vannucci RC, Brierley JB 1981 The influence of immaturity on
hypoxic-ischemic brain damage in the rat. Ann Neurol 9:131–141
14. de Medinaceli L, Freed WJ, Wyatt RJ 1982 An index of the functional condition of
rat sciatic nerve based on measurements made from walking tracks. Exp Neurol
77:634–643
15. Ivacko JA, Sun R, Silverstein FS 1996 Hypoxic-ischemic brain injury induces an
acute microglial reaction in perinatal rats. Pediatr Res 39:39–47
16. Wang X, Karlsson JO, Zhu C, Bahr BA, Hagberg H, Blomgren K 2001 Caspase-3
activation after neonatal rat cerebral hypoxia-ischemia. Biol Neonate 79:172–179
17. Hu BR, Liu CL, Ouyang Y, Blomgren K, Siesjo BK 2000 Involvement of caspase-3
in cell death after hypoxia-ischemia declines during brain maturation. J Cereb Blood
Flow Metab 20:1294–1300
18. Adams TE, Bodmer JG, Bodmer WF 1983 Production and characterization of
monoclonal antibodies recognizing the alpha-chain subunits of human ia alloanti-
gens. Immunology 50:613–624
19. Hare GM, Evans PJ, Mackinnon SE, Best TJ, Midha R, Szalai JP, Hunter DA 1993
Walking track analysis: utilization of individual footprint parameters. Ann Plast Surg
30:147–153
20. Bona E, Andersson AL, Blomgren K, Gilland E, Puka-Sundvall M, Gustafson K,
Hagberg H 1999 Chemokine and inflammatory cell response to hypoxia-ischemia in
immature rats. Pediatr Res 45:500–509
21. Daley GQ, Goodell MA, Snyder EY 2003 Realistic prospects for stem cell thera-
peutics. Hematology (Am Soc Hematol Educ Program) 1:398–418
22. Escolar ML, Poe MD, Provenzale JM, Richards KC, Allison J, Wood S, Wenger DA,
Pietryga D, Wall D, Champagne M, Morse R, Krivit W, Kurtzberg J 2005 Trans-
plantation of umbilical-cord blood in babies with infantile Krabbe’s disease. N Engl
J Med 352:2069–2081
23. Muramatsu K, Fukuda A, Togari H, Wada Y, Nishino H 1997 Vulnerability to
cerebral hypoxic-ischemic insult in neonatal but not in adult rats is in parallel with
disruption of the blood-brain barrier. Stroke 28:2281–2289
24. Park KI, Liu S, Flax JD, Nissim S, Stieg PE, Snyder EY 1999 Transplantation of
neural progenitor and stem cells: developmental insights may suggest new therapies
for spinal cord and other CNS dysfunction. J Neurotrauma 16:675–687
25. Risdon G, Gaddy J, Broxmeyer HE 1994 Allogeneic responses of human umbilical
cord blood. Blood Cells 20:566–572
26. Rainsford E, Reen DJ 2002 Interleukin 10, produced in abundance by human
newborn T cells, may be the regulator of increased tolerance associated with cord
blood stem cell transplantation. Br J Haematol 116:702–709
27. Zigova T, Song S, Willing AE, Hudson JE, Newman MB, Saporta S, Sanchez-Ramos J,
Sanberg PR 2002 Human umbilical cord blood cells express neural antigens after
transplantation into the developing rat brain. Cell Transplant 11:265–274
28. Balsam LB, Wagers AJ, Christensen JL, Kofidis T, Weissman IL, Robbins RC 2004
Haematopoietic stem cells adopt mature haematopoietic fates in ischaemic myocar-
dium. Nature 428:668–673
29. Ourednik J, Ourednik V, Lynch WP, Schachner M, Snyder EY 2002 Neural stem
cells display an inherent mechanism for rescuing dysfunctional neurons. Nat Bio-
technol 20:1103–1110
30. Lu P, Jones LL, Snyder EY, Tuszynski MH 2003 Neural stem cells constitutively
secrete neurotrophic factors and promote extensive host axonal growth after spinal
cord injury. Exp Neurol 181:115–129
249UMBILICAL CELLS REDUCE SPASTIC PARESIS
... Twenty-five studies evaluated apoptosis using the markers of caspase 3 (n = 13) or TUNEL (n = 13), with one study evaluating both. [46] Seven studies were excluded from the meta-analysis due to lack of quantitative data available or brain structure not classified as grey or white matter [44,45,53,56,65,68,75]. As evident in Figure 2E,F, the 18 studies included in the meta-analysis were further grouped into studies that assessed grey matter and white matter brain regions. ...
... Twenty-two papers assessed Iba-1 (n = 12) or ED-1 (n = 10) as a marker of microglial activation in grey matter brain regions and 6 papers assessed this in white matter brain regions. Due to unavailable data for analysis or brain regions not being classified as grey or white matter, 6 papers were excluded for meta-analysis for microglial activation in grey matter [24,30,43,53,62,68], and no papers excluded for white matter. As shown in Figure 3C,D, there were 23 study entries and 8 study entries in the forest plots, respectively. ...
Article
Full-text available
Perinatal brain injury is a major contributor to long-term adverse neurodevelopment. There is mounting preclinical evidence for use of umbilical cord blood (UCB)-derived cell therapy as potential treatment. To systematically review and analyse effects of UCB-derived cell therapy on brain outcomes in preclinical models of perinatal brain injury. MEDLINE and Embase databases were searched for relevant studies. Brain injury outcomes were extracted for meta-analysis to calculate standard mean difference (SMD) with 95% confidence interval (CI), using an inverse variance, random effects model. Outcomes were separated based on grey matter (GM) and white matter (WM) regions where applicable. Risk of bias was assessed using SYRCLE, and GRADE was used to summarise certainty of evidence. Fifty-five eligible studies were included (7 large, 48 small animal models). UCB-derived cell therapy significantly improved outcomes across multiple domains, including decreased infarct size (SMD 0.53; 95% CI (0.32, 0.74), p < 0.00001), apoptosis (WM, SMD 1.59; 95%CI (0.86, 2.32), p < 0.0001), astrogliosis (GM, SMD 0.56; 95% CI (0.12, 1.01), p = 0.01), microglial activation (WM, SMD 1.03; 95% CI (0.40, 1.66), p = 0.001), neuroinflammation (TNF-α, SMD 0.84; 95%CI (0.44, 1.25), p < 0.0001); as well as improved neuron number (SMD 0.86; 95% CI (0.39, 1.33), p = 0.0003), oligodendrocyte number (GM, SMD 3.35; 95 %CI (1.00, 5.69), p = 0.005) and motor function (cylinder test, SMD 0.49; 95 %CI (0.23, 0.76), p = 0.0003). Risk of bias was determined as serious, and overall certainty of evidence was low. UCB-derived cell therapy is an efficacious treatment in pre-clinical models of perinatal brain injury, however findings are limited by low certainty of evidence.
... Multiple studies have demonstrated that umbilical cord blood cells protect the brain in HIE and NAIS. [123][124][125][126] Currently, it is unknown which cell types present in the umbilical cord blood are necessary for the beneficial effects observed in these models. Saha et al. 127 demonstrated that CD14 + monocytes from the umbilical cord blood protected neurons and reduced glial activation in organotypic forebrain slice cultures subjected to oxygen-glucose deprivation. ...
Article
Full-text available
Key points Monocytes are recruited to the brain within the first days following neonatal arterial ischemic stroke (NAIS) and hypoxic‐ischemic encephalopathy (HIE). More severe lesions or a combination of HIE and systemic inflammatory insults lead to more robust recruitment of monocytes to the brain. Monocytes differentiate into macrophages and microglia‐like cells, which can remain in the brain for several months following NAIS and HIE. CCR2 signaling is critical for monocyte infiltration and represents a potential therapeutic target. The outcomes of CCR2 inhibition or knockout vary depending on the HIE model used, highlighting the need for more studies in different models. Models of cerebral ischemia in neonates and adults indicate that monocytes may contribute to both brain damage, through their pro‐inflammatory activity and ability to modify perineuronal nets, and brain repair and functional recovery, through efferocytosis and their role in vascular repair. Umbilical cord blood monocytes and preconditioned monocytes could be used for the development of cell‐based therapies. Pharmacological interventions targeting monocyte infiltration or function require further testing in experimental models of HIE and NAIS.
... Therefore, modulation of microglial phenotype, immunomodulation, or repair of microglial function may be a novel therapeutic strategy for the treatment of neurological disorders accompanied by inflammation. Umbilical cord blood contains hematopoietic stem cells, endothelial progenitor cells, and mesenchymal stromal cells (MSCs) that alter brain connectivity and modulate inflammation 37,38 . The infusion of autologous umbilical cord blood cells (UCBs) has been shown to be safe in individuals with cerebral palsy (CP), ASD, and other acquired brain injuries. ...
Article
Full-text available
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder characterized by difficulties in social communication, repetitive behaviors, and restricted interests, with onset early in life. The prevalence of ASD has increased worldwide in the last two decades. However, there is currently no effective therapy for ASD. Therefore, it is important to develop new strategies for ASD treatment. Evidence for the relationship between ASD and neuroinflammation, ASD and microglia, and ASD and glucose metabolism has increased rapidly in recent decades. We reviewed 10 clinical studies on cell therapies for individuals with ASD. Almost all studies showed good outcomes and no remarkable adverse events. Over the past decades, the neurophysiological characteristics of ASD have been shown to be impaired communication, cognition, perception, motor skills, executive function, theory of mind, and control of emotions. Recent studies have focused on the roles of immune pathology, such as neuroinflammation, microglia, cytokines, and oxidative stress, in ASD. We also focused on glucose metabolism in patients with ASD. The significance of gap junction-mediated cell-cell interactions between the cerebral endothelium and transplanted cells was observed in both bone marrow mononuclear cells and mesenchymal stromal cells transplantation. Owing to the insufficient number of samples, cell therapies, such as umbilical cord blood cells, bone marrow mononuclear cells, and mesenchymal stromal cells, will be a major challenge for ASD. As a result of these findings, a new paradigm for cell therapy for autism may emerge.
... Kolejne prace badawcze z użyciem mo deli zwierzęcych uwidoczniły potencjał neuroprotekcyjny komórek UCB w okołoporodowym uszkodzeniu mózgu u szczurów w przypadku gdy komórki UCB zostały po dane w ciągu pierwszych 24 godzin po urazie niedotlenie niowoniedokrwiennym. Poprawie ulegała kontrola moto ryczna, wzorce chodzenia i niedowład spastyczny [21][22][23]. Odnotowano również zmniejszenie degeneracji neuronal nej, apoptozy i aktywacji mikrogleju [24]. W ostatnich la tach wykazano, że MSC pochodzące z UCB mogą zwięk szyć neuroprotekcyjne korzyści hipotermii u noworodków szczurów narażonych na niedotlenienie i niedokrwienie -co jest szczególnie istotne, biorąc pod uwagę, że hipoter mia terapeutyczna lecznicza jest obecnie standardem opie ki nad noworodkami z encefalopatią niedotlenieniowonie dokrwienną (HIE, ang. ...
Article
Hypoxic-ischemic encephalopathy (HIE) is a leading cause of long-term neurological disability in neonates and adults. Despite emerging advances in supportive care, like the most effective approach, hypothermia, poor prognosis has still been present in current clinical treatment for HIE. Stem cell therapy has been adopted for treating cerebral ischemia in preclinical and clinical trials, displaying its promising therapeutic value. At present, reported treatments for stroke employed stem cells to replace the lost neurons and integrate them into the existing host circuitry, promoting the release of growth factors to support and stimulate endogenous repair processes, etc. In this review, a meaningful overview to numerous studies published up to now was presented by introducing the preclinical and clinical research status of stem cell therapy for cerebral ischemia and hypoxia, discussing potential therapeutic mechanisms of stem cell transplantation for curing HI-induced brain injury, summarizing a series of approaches for marking transplanted cells and existing imaging systems for stem cell labeling and in vivo tracking, and expounding the endogenous regeneration capability of stem cells in the newborn brain when subjected to an HI insult. Additionally, it's promising to combine stem therapy with neuromodulation through specific regulation of neural circuits. The crucial neural circuits across different brain areas related to functional recovery are of great significance for the application of neuromodulation strategies after the occurrence of neonatal hypoxic ischemic encephalopathy (NHIE).
Article
Full-text available
Introduction We have previously described preclinical literature which supports umbilical cord blood-derived cell (UCBC) therapy as an efficacious treatment for perinatal brain injury. However, efficacy of UCBCs may be influenced by different patient population and intervention characteristics. Objectives To systematically review the effects of UCBCs on brain outcomes in animal models of perinatal brain injury across subgroups to better understand the contribution of model type (preterm versus term), brain injury type, UCB cell type, route of administration, timing of intervention, cell dosage, and number of doses. Methods A systematic search of MEDLINE and Embase databases was performed to identify studies using UCBC therapy in animal models of perinatal brain injury. Subgroup differences were measured by chi2 test where possible. Results Differential benefits of UCBCs were seen across a number of subgroup analyses including intraventricular hemorrhage (IVH) vs. hypoxia ischemia (HI) model (apoptosis white matter (WM): chi2 = 4.07; P = .04, neuroinflammation-TNF-α: chi2 = 5.99; P = .01), UCB-derived mesenchymal stromal cells (MSCs) vs. UCB-derived mononuclear cells (MNCs) (oligodendrocyte WM: chi2 = 5.01; P = .03, neuroinflammation-TNF-α: chi2 = 3.93; P = .05, apoptosis grey matter (GM), astrogliosis WM), and intraventricular/intrathecal vs. systemic routes of administration (microglial activation GM: chi2 = 7.51; P = .02, astrogliosis WM: chi2 = 12.44; P = .002). We identified a serious risk of bias and overall low certainty of evidence. Conclusions Preclinical evidence suggests UCBCs to show greater efficacy in the injury model of IVH compared to HI, the use of UCB-MSCs compared to UCB-MNCs and the use of local administrative routes compared to systemic routes in animal models of perinatal brain injury. Further research is needed to improve certainty of evidence and address knowledge gaps.
Chapter
Cerebral Palsy and the developmental disabilities have their origins from birth and last a lifetime, leading to tremendous financial and emotional burdens on society and the families. The etiology for the majority of causes of cerebral palsy and developmental disability arises from before the time of birth. It can be attributed in a good percentage of cases to intra-uterine hypoxia-ischemia, perinatal asphyxia, or fetal inflammation, and less often to underlying genetic conditions (though increasingly so), toxins during pregnancy, or trauma. Treatment for these causes and ultimate outcomes has been limited to date. Whereas therapeutic hypothermia post-ischemically has been introduced as standard of care in recent years, few other therapies have been accepted. Importantly several studies have now affirmed theutilization of therapies prior to the time of birth, which is important in reducing, not only the degree of injury, but perhaps the onset of the injury and damage altogether. In addition, sophisticated approaches to brain regeneration in the form of transcutaneous magnetic stimulation and stem cell therapy have made significant strides in both pre-clinical and clinical studies. The current chapter is not an attempt at being all-inclusive with respect to therapeutic interventions, but rather provide an introduction into those accepted therapies and several that can be looked forward to in the near future.
Article
Full-text available
Cerebral palsy (CP), the most common disability in childhood, is adevastating non-progressive ailment of the infants' brain with lifelongsequelae, e.g., spastic paresis, chronic pain, inability to walk, intellectualdisability, behavioral disorders, for which there is no cure at present.CP may develop after pediatric brain damage caused, e.g., by hypoxic-ischemia, periventricular leukomalacia, intracranial hemorrhage, hypoxic-ischemicencephalopathy, trauma, stroke, and infection. About 17 million peopleworldwide live with cerebral palsy as a result of pediatric brain damage.This reflects both the magnitude of the personal, medical, andsocioeconomic global burden of this brain disorder and the overt unmettherapeutic needs of the pediatric population.This review will focus on recent preclinical, clinical, and regulatorydevelopments in cell therapy for infantile cerebral palsy bytransplantation of cord blood derived mononuclear cells from bench tobedside. The body of evidence suggests that cord blood cell therapy ofcerebral palsy in the autologous setting is feasible, effective, and safe,however, adequately powered phase 3 trials are overdue. https://mc.manuscriptcentral.com/jpmedJournal of Perinatal Medicine https://www.researchgate.net/publication/365303033_2022_10_27_JPMed20220505_Accept_in_its_present_form [accessed Nov 11 2022].
Article
Full-text available
In the last two decades, the field of stem cell neurobiology has become one of the hottest, most rapid growing arenas in neuroscience. When, as a very young scientist, I entered the field of progenitor biology under the guidance of Professor Pasquale Graziadei at Florida State University, thoughts about neuronal turnover and migration of cells were not often the subjects of discussion. The existence of “dreaming pools” of immature cells that could serve as replacements for existing neuronal populations within the brains of either birds or rodents became obvious much later. But beginning in the early 1990s, knowledge in this field grew like an avalanche. Today, no one doubts the existence of stem/progenitor cells within the brain, and scientists, like enthusiastic prospectors, have turned their attention to the next level, exploring the properties of these valuable cell sources and developing methods to utilize them in various therapeutic treatments. The development of therapeutic treatments, however, requires extensive and thorough knowledge of the properties and characteristics of these cells within their natural brain environment. This book, Stem Cells and CNS Development, edited by Mahendra Rao, has appeared exactly at the right time to provide this essential information.
Article
Full-text available
The knowledge on fetal and neonatal circulatory physiology accumulated by basic scientists and clinicians over the years has contributed considerably to the recent decline of perinatal morbidity and mortality. This review will summarize the peculiarities of the fetal circulation, the distribution of organ blood flow during normoxemia, and that during oxygen lack caused by various experimental perturbations. Furthermore, the relation between oxygen delivery and tissue metabolism during oxygen lack as well as evidence to support a new concept will be presented along with the principal cardiovascular mechanisms involved. Finally, blood flow and oxygen delivery to the principal fetal organs will be examined and discussed in relation to organ function. The fetal circulatory response to hypoxemia and asphyxia is a centralization of blood flow in favour of the brain, heart, and adrenals and at the expense of almost all peripheral organs, particularly of the lungs, carcass, skin and scalp. This response is qualitatively similar but quantitatively different under various experimental conditions. However, at the nadir of severe acute asphyxia the circulatory centralization cannot be maintained. Then there is circulatory decentralization, and the fetus will experience severe brain damage if not expire unless immediate resuscitation occurs. Future work in this field will have to concentrate on the important questions, what factors determine this collapse of circulatory compensating mechanisms in the fetus, how does it relate to neuronal damage, and how can the fetal brain be pharmacologically protected against the adverse effects of asphyxia.
Article
Studies of the regenerating hematopoietic system have led to the definition of many of the fundamental principles of stem cell biology. Therapies based on a range of tissue stem cells have been widely touted as a new treatment modality, presaging an emerging new specialty called regenerative medicine that promises to harness stem cells from embryonic and somatic sources to provide replacement cell therapies for genetic, malignant, and degenerative conditions. Insights borne from stem cell biology also portend development of protein and small molecule therapeutics that act on endogenous stem cells to promote repair and regeneration. Much of the newfound enthusiasm for regenerative medicine stems from the hope that advances in the laboratory will be followed soon thereafter by breakthrough treatments in the clinic. But how does one sort through the hype to judge the true promise? Are stem cell biologists and the media building expectations that cannot be met? Which diseases can be treated, and when can we expect success? In this review, we outline the realms of investigation that are capturing the most attention, and consider the current state of scientific understanding and controversy regarding the properties of embryonic and somatic (adult) stem cells. Our objective is to provide a framework for appreciating the promise while at the same time understanding the challenges behind translating fundamental stem cell biology into novel clinical therapies.
Article
Summary: Background: Cerebral hypothermia can improve outcome of experimental perinatal hypoxia-ischaemia. We did a multicentre randomised controlled trial to find out if delayed head cooling can improve neurodevelopmental outcome in babies with neonatal encephalopathy. Methods: 234 term infants with moderate to severe neonatal encephalopathy and abnormal amplitude integrated electroencephalography (aEEG) were randomly assigned to either head cooling for 72 h, within 6 h of birth, with rectal temperature maintained at 34–35°C (n=116), or conventional care (n=118). Primary outcome was death or severe disability at 18 months. Analysis was by intention to treat. We examined in two predefined subgroup analyses the effect of hypothermia in babies with the most severe aEEG changes before randomisation—ie, severe loss of background amplitude, and seizures—and those with less severe changes. Findings: In 16 babies, follow-up data were not available. Thus in 218 infants (93%), 73/110 (66%) allocated conventional care and 59/108 (55%) assigned head cooling died or had severe disability at 18 months (odds ratio 0·61; 95% CI 0·34–1·09, p=0·1). After adjustment for the severity of aEEG changes with a logistic regression model, the odds ratio for hypothermia treatment was 0·57 (0·32–1·01, p=0·05). No difference was noted in the frequency of clinically important complications. Predefined subgroup analysis suggested that head cooling had no effect in infants with the most severe aEEG changes (n=46, 1·8; 0·49–6·4, p=0·51), but was beneficial in infants with less severe aEEG changes (n= 172, 0·42; 0·22–0·80, p=0·009). Interpretation: These data suggest that although induced head cooling is not protective in a mixed population of infants with neonatal encephalopathy, it could safely improve survival without severe neurodevelopmental disability in infants with less severe aEEG changes.
Article
Two monoclonal antibodies, TAL-1B5 and TAL-3C3, specific for human Ia alpha-chain subunits have been produced by fusing P3/NSI/1-Ag4-1 mouse myeloma cells with spleen cells from a BALB/c mouse immunized with purified alpha-chains. Specificity for the alpha-chain subunits was initially established using a solid-phase radioimmunoassay. Indirect binding assays demonstrated that TAL-1B5 bound strongly to all human B lymphoblastoid lines tested and to CLLs, but only weakly to PBL-B cells and not to PBL-T cells or the T-cell lines Molt 4 and HSB-2. TAL-3C3 bound only weakly to B lymphoblastoid lines and not to CLLs or PBL-B cells. From 125I cell surface-labelled lysates TAL-1B5 immunoprecipitated a 33,000(alpha):28,000(beta) Ia dimer, but TAL-3C3 failed to immunoprecipitate cell surface molecules. Under denaturing conditions, however, both TAL-1B5 and TAL-3C3 immunoprecipitated the 33,000 alpha-chain subunit. Competitive inhibition studies demonstrated that both monoclonal antibodies recognize the same or spatially related alpha-chain antigenic determinants with some slight cross-reactivity against beta-chains. 2D-NEPHGE/SDS-PAGE analysis of TAL-1B5 immunoprecipitates from [35S]-methionine biosynthetically labelled cells revealed the presence of a number of alpha-chain spots in association with beta-chain products of three previously described loci (beta-1, beta-2, beta-3) suggesting that this antibody recognizes an antigenic site common to those human Ia alpha-chains so far identified.
Article
There is no widely accepted quantitative method for evaluating the functional effects of peripheral nerve damage in animals. In the present study, a method for evaluating sciatic nerve damage was developed from measurements of the prints of the hind feet of walking rats preserved on X-ray film. Four variables were measured from these tracks, and comparisons between the damaged (experimental) and intact (normal) side were converted to percent deficits and averaged to obtain a “sciatic functional index” (SFI). The SFI was then measured under normal conditions, after nerve transection, nerve crush, and sham procedures. Reliability and repeatability of the SFI were found to be excellent. The effects of sciatic nerve transection and nerve crush evaluated by this method agreed very well with other methods of evaluating nerve damage. We conclude that the SFI provides a simple, accurate, reliable, and repeatable method for evaluating the functional condition of sciatic nerve in rats.
Article
Brain damage in the Levine preparation (unilateral common carotid artery ligation with hypoxia) consists of ischemic neuronal alterations in the ipsilateral forebrain. As the model has been restricted to adult animals, unilateral common carotid artery ligation was carried out in 7-day-postnatal rats. Four to 8 hours later the 25 pups were exposed to 8% oxygen at 37°C for 3.5 hours. Controls consisted of littermates subjected to carotid ligation without subsequent hypoxia, hypoxia without prior ligation, and neither ligation nor hypoxia. After hypoxia the animals were returned to their dams and appeared normal for up to 50 hours. All pups were then killed by perfusion-fixation. Moderate to severe ischemic neuronal changes were seen in the ipsilateral cerebral cortex, striatum, and hippocampus in at least 90% of the animals and included infarction in 56% of the brains. Cortical damage was occasionally laminar but more often occurred in columns at right angles to the pial surface. Unlike adult animals, there was necrosis of white matter, greater ipsilaterally, originating in and spreading from myelinogenic foci. The evolution of ischemic cell change and the associated gliomesodermal reaction was more rapid than in the adult. In 22 additional pups subjected to carotid artery ligation and hypoxia, brains were analyzed for water content. Significant increases (0.6 to 3.3%) in water content of the ipsilateral hemispheres occurred in 11 of 22 brains (50%). Unilateral ischemia combined with hypoxia in developing rats therefore results in neuronal destruction in the same brain regions as in adult animals, but also causes necrosis of white matter. The incidence of increased water content was similar to that of overt infarction. Thus, as previously shown in the adult, brain edema is a consequence rather than a cause of major ischemic damage in the immature animal.
Article
Human umbilical cord blood (CB) is increasingly used as an alternative to bone marrow as a source of stem and progenitor cells for pediatric patients. We have evaluated the alloreactive potential of cord blood T cells in vitro. Our findings demonstrate that in a primary mixed leukocyte culture CB T cells demonstrate strong proliferative responses to allogeneic stimulation but little to no generation of cytotoxic effector function. Furthermore, restimulation of primary cultures results in a state of proliferative unresponsiveness. Such diminished cytotoxic and proliferative responses may, in part, be related to the low incidence of graft vs. host disease thus far noted in human CB transplants.
Article
In a model of perinatal hypoxic-ischemic brain damage, we examined the neuroprotective efficacy of posttreatment with the NMDA receptor antagonist MK-801 and the AMPA receptor antagonist NBQX. Unilateral brain damage developed in 95% of rat pups subjected to hypoxia-ischemia with a 27.8 +/- 1.2% weight deficit of the damaged hemisphere. MK-801 in doses of 0.3 and 0.5 mg/kg i.p. reduced the brain damage by 61% (p < 0.001) and 43% (p < 0.001), respectively. A higher dose of MK-801 (0.75 mg/kg) did not offer neuroprotection. Treatment with NBQX (40 mg/kg) reduced the hemispheric lesion by 28% (p < 0.05). In conclusion, posttreatment with both NBQX and low doses of MK-801 reduced perinatal brain damage. The NMDA receptor antagonist offered stronger neuroprotection which is in agreement with a proposed NMDA receptor hyperactivity around postnatal day 7 in rats.