ArticlePDF Available

Estradiol attenuates hyperoxia-induced cell death in the developing white matter

Authors:

Abstract and Figures

Periventricular leukomalacia is the predominant type of brain injury in preterm infants underlying the development of cerebral palsy. Periventricular leukomalacia has its peak incidence at 23 to 32 weeks postconceptional age characterized by extensive oligodendrocyte migration and maturation. Oxygen toxicity has been identified as a possible contributing factor to the pathogenesis of cerebral palsy in survivors of preterm birth. 17beta-estradiol (E2) is important for the development and function of the central nervous system. Furthermore, neuroprotective properties have been attributed to estrogens. We examined the effect of E2 on hyperoxia-induced cell death in the developing white matter in the rat brain. Six-day-old (P6) rat pups, the immature oligodendroglial cell line (OLN-93), and primary oligodendrocyte cultures were subjected to 80% O(2) in the presence or absence of E2 (600 microg/kg intraperitoneally in vivo, 10(-6)-10(-10)M in vitro). Cell counts and lactate dehydrogenase assay were used to assess cell survival. Immunoblot analysis was used for detection of estrogen receptor expression and investigation of apoptotic signaling pathways. White matter injury was assessed by myelin basic protein immunocytochemistry at P11. E2 produced significant dose-dependent protection against oxygen-induced apoptotic cell death in primary oligodendrocytes. Treatment with E2 prevented hyperoxia-induced proapoptotic Fas-upregulation and caspase-3 activation. Finally, E2 antagonized hyperoxia-induced inactivation of extracellular signal-regulated kinase 1 and 2 and Akt, key kinases of the mitogen-activated protein kinase and phosphatidylinositol 3-kinase cell survival promoting pathways, respectively. Loss of myelin basic protein labeling was seen in P11 pups after oxygen exposure, and E2 attenuated this injury. These results suggest a possible role for estrogens in the prevention of neonatal oxygen-induced white matter injury.
A) Effect of 17β-estradiol (E2) on apoptosis signaling proteins in immature oligodendrocytes (OLN-93) exposed to hyperoxia. Immunoblot densitometric quantification of protein levels for Fas shows increased Fas-expression after 6 and 12 hours of hyperoxia. ***p < 0.001, **p < 0.01, comparison between normoxia (0 hours) or oxygen exposure for 2, 6, 12, or 24 hours (analysis of variance). Treatment with estrogen (100nM) eliminates Fas-upregulation. Blot is representative of a series of blots of three independent experiments. Graphs represent means ± standard error of means (SEMs) of the density ratios of Fas to β-actin. #p < 0.05, comparison between vehicle and E2-treated cells, Student's t test. (B) Fas messenger RNA (mRNA) expression in P7-day-old whole rat brains 12 or 24 hours after 80% oxygen exposure and in P6-day-old whole rat brains treated with E2 before hyperoxia. There was an increase in mRNA levels for Fas 12 and 24 hours after hyperoxia. Treatment of P6-day-old rats with E2 before hyperoxia exposure attenuated Fas upregulation in comparison with Fas-mRNA levels of untreated littermates. The results of densitometric analysis of the gels are presented in reference to 18S ribosomal RNA (rRNA). Data represent the ratio (%) of the density of the Fas band to the 18S rRNA band ± SEM (n = 4). *p < 0.05, comparison between P7-day-old whole rat brains 12 or 24 hours after 80% oxygen exposure and P7-day-old whole rat brains treated with E2 before hyperoxia (analysis of variance). (C, D) Hyperoxia triggered an increase of Fas protein after 12 and 24 hours in lysates from P7-day-old whole rat brain (WB) and white matter preparation (WM). Treatment of P6-day-old rats with E2 (600μg/kg intraperitoneally) before hyperoxia exposure attenuated Fas upregulation in comparison with Fas levels of untreated littermates. Blots are representative of a series of four blots. Densitometric quantification of the blots is presented. Columns represent means ± SEMs of the density ratios of Fas to β-actin (n = 4 per group). ***p < 0.001, comparison between vehicle and E2, Student's t test.
… 
Content may be subject to copyright.
Estradiol Attenuates Hyperoxia-Induced Cell
Death in the Developing White Matter
Bettina Gerstner, MD,
1,2
Marco Sifringer, MSc,
1,3
Mark Dzietko, MD,
1
Alexandra Schu¨ller, MD,
1
Joan Lee, BA,
2
Sinno Simons, MD,
1
Michael Obladen, MD, PhD,
1
Joseph J. Volpe, MD,
2
Paul A. Rosenberg, MD, PhD,
2
and Ursula Felderhoff-Mueser, MD, PhD
1
Objective: Periventricular leukomalacia is the predominant type of brain injury in preterm infants underlying the development
of cerebral palsy. Periventricular leukomalacia has its peak incidence at 23 to 32 weeks postconceptional age characterized by
extensive oligodendrocyte migration and maturation. Oxygen toxicity has been identified as a possible contributing factor to the
pathogenesis of cerebral palsy in survivors of preterm birth. 17-estradiol (E2) is important for the development and function
of the central nervous system. Furthermore, neuroprotective properties have been attributed to estrogens. We examined the effect
of E2 on hyperoxia-induced cell death in the developing white matter in the rat brain.
Methods: Six-day-old (P6) rat pups, the immature oligodendroglial cell line (OLN-93), and primary oligodendrocyte cultures
were subjected to 80% O
2
in the presence or absence of E2 (600g/kg intraperitoneally in vivo, 10
6
–10
10
M in vitro). Cell
counts and lactate dehydrogenase assay were used to assess cell survival. Immunoblot analysis was used for detection of estrogen
receptor expression and investigation of apoptotic signaling pathways. White matter injury was assessed by myelin basic protein
immunocytochemistry at P11.
Results: E2 produced significant dose-dependent protection against oxygen-induced apoptotic cell death in primary oligoden-
drocytes. Treatment with E2 prevented hyperoxia-induced proapoptotic Fas-upregulation and caspase-3 activation. Finally, E2
antagonized hyperoxia-induced inactivation of extracellular signal-regulated kinase 1 and 2 and Akt, key kinases of the mitogen-
activated protein kinase and phosphatidylinositol 3-kinase cell survival promoting pathways, respectively. Loss of myelin basic
protein labeling was seen in P11 pups after oxygen exposure, and E2 attenuated this injury.
Interpretation: These results suggest a possible role for estrogens in the prevention of neonatal oxygen-induced white matter
injury.
Ann Neurol 2007;61:562–573
Advances in neonatal intensive care have markedly im-
proved survival rates of premature infants. Unfortu-
nately, a substantial proportion of very-low-birth-
weight infant survivors have neurological deficits that
affect motor and cognitive function.
1,2
Long-term neu-
rocognitive impairment restricts quality of life for af-
fected individuals and their families and poses a con-
siderable socioeconomic problem.
3
The neuropathology of perinatal brain injury is com-
plex and involves gray and white matter structures to
varying degrees, depending on the gestational age and
the developmental stage.
4,5
The timing of vulnerability
coincides with the peak of the brain growth spurt,
which starts at about midpregnancy in humans and ex-
tends into the third postnatal year.
6
Periventricular leu-
komalacia (PVL) is the predominant type of injury in
preterm infants and leads to a chronic deficit of white
matter structures. It has its peak incidence during a
well-defined period in human brain development
(23–32 weeks, postconceptional age) characterized by
extensive oligodendrocyte (OL) migration and matura-
tion.
7
Oxygen is widely used in neonatal intensive care.
However, it is implicated in the pathogenesis of neo-
natal chronic lung disease and retinopathy of the pre-
maturity. Our group has recently shown that hyperoxia
is a powerful trigger for widespread apoptotic neuronal
death in the developing brain. Hyperoxia increased the
density of degenerating cells in various brain regions of
7-day-old Wistar rats and C57/BL6 mice such as the
caudate nucleus, layers II and IV of the frontal, pari-
etal, cingulate, and retrosplenial cortices, as well as
From the
1
Department of Neonatology, Charite´ Campus Virchow-
Klinikum, Berlin, Germany;
2
Department of Neurology and Neu-
robiology Program, Children’s Hospital Boston, Boston, MA; and
3
Department of Pediatric Neurology, Children’s Hospital, Medical
Faculty Carl Gustav Carus, Technical University Dresden, Dresden,
Germany.
P.A.R. and U.F.-M. contributed equally to this work.
Received Dec 7, 2006, and in revised form Feb 2, 2007. Accepted
for publication Feb 9, 2007.
Published online Apr 11, 2007, in Wiley InterScience
(www.interscience.wiley.com). DOI: 10.1002/ana.21118
Address correspondence to Dr Gerstner, Department of Neonatol-
ogy, Campus Virchow Klinikum, Universita¨tsmedizin Berlin,
Charite´, Augustenburger Platz 1, 13353 Berlin, Germany.
E-mail: bettina.gerstner@charite.de
562 © 2007 American Neurological Association
Published by Wiley-Liss, Inc., through Wiley Subscription Services
white matter tracts and the periventricular region.
8
This cell death is associated with oxidative stress, de-
creased expression of neurotrophins, decreased activa-
tion of neurotrophin-regulated pathways,
8
and in-
creased levels of proinflammatory cytokines.
9
Recently,
we investigated the effects of hyperoxia on cultured rat
precursor, immature and mature oligodendroglia cells,
derived from the permanent oligodendrocyte cell line
(OLN-93). Hyperoxia initiated the apoptotic cascade
in immature OLs and preoligodendroglial cells, but not
in mature OLs in vitro.
10
New pharmacological ap-
proaches are necessary to limit the neurotoxic effects of
hyperoxia in the developing brain.
17-estradiol (E2) is one promising candidate for
neuroprotection. E2 and the estrogen receptor (ER)
play important roles in the development and function
of the central nervous system.
11,12
Its receptors, ER-
and ER-show a specific distribution with high den-
sities around the ventricles, the region where brain cell
progenitors are generated.
13
Neuroprotective effects of
E2 in the mature brain have been observed in several
models of neurotoxicity involving hypoxia-ischemia
and excitotoxicity.
14,15
Female steroid hormones can
influence the apoptotic cascade at different stages.
16
Moreover, E2 has antioxidative properties that lead to
the reduction of free radicals.
17
E2 also has profound
effects on the function and plasticity of the brain and
proliferation, differentiation, and migration of neurons
are controlled by E2.
18
During pregnancy, E2 levels increase up to 100-fold
in the placenta.
19
The fetus is also exposed to these
increasing hormone levels. At birth, after the umbilical
cord is clamped, the levels of E2 decrease by a factor of
100 within 24 hours.
19
Premature infants experience
this hormone deprivation and simultaneous increase of
the oxgygen tissue tension much earlier than infants
born at term.
In an attempt to characterize measures that will
counteract injury to the developing white matter, this
study focused on the effect of E2 on premyelinating
oligodendrocytes (pre-OL) in established models of ap-
optotic brain damage in vitro and in vivo.
Materials and Methods
Cell Cultures
OLIGODENDROGLIA CELL LINE. The OLN-93 cell line
derived from spontaneously transformed cells in primary rat
brain glia culture was kindly provided by Dr C. Richter-
Landsberg (Institute of Molecular Neurobiology, Oldenburg,
Germany).
20
OLN-93 cells bear the morphological and an-
tigenic properties of 5- to 10-day-old (postnatal time) cul-
tured rat brain OLs. These resemble the intermediate stage
between the pre-OL (O4O1
MBP
) and the mature OL
(O4
O1
MBP
). Cells were maintained in Dulbecco’s
minimum essential medium, with 3.7gm/L NaHCO
3
,
25mM HEPES
3
, 4.5gm/L D-glucose, 4.4gm/L NaCl, con-
taining heat-inactivated 10% fetal calf serum. Monolayers
were cultured at 37°C in a humidified, 5% CO
2
atmosphere
with medium replenishment every 2 to 3 days.
OLIGODENDROCYTE PRIMARY CULTURES. Primary rat
OLs were prepared from the cerebral hemispheres of
Sprague–Dawley rats at postnatal days 1 to 2 using a shaking
method
21
with modifications, as described previously.
22
An-
imals were killed by decapitation. Purified OLs were cultured
for 7 to 8 days in a serum-free basal-defined medium: Dul-
becco’s minimum essential medium, 0.1% bovine serum al-
bumin, 50g/ml apo-transferrin, 50g/ml insulin, 30nM so-
dium selenite, 10nM D-biotin, 10nM hydrocortisone,
200ML-cystine, 10ng/ml platelet-derived growth factor,
and 10ng/ml basic fibroblast growth factor. At 7 to 8 days,
the cultures were composed primarily of progenitors and pre-
OLs (O4
O1
MBP
). The purity of OL cultures was con-
sistently greater than 95% OLs with less than 5% astrocyte
contamination. Primary OL cultures and OLN-93 cells were
subjected to 80% O
2
in the presence or absence of E2
(10
6
–10
10
M; Sigma, St. Louis, MO). E2 was given at 0,
2, 4, 6, 12, 24, and 48 hours before oxygen exposure.
IMMUNOCYTOCHEMISTRY. Cells were fixed with 4%
paraformaldehyde in phosphate-buffered saline (PBS) for 10
minutes at room temperature (RT), washed 3 times with
PBS, and blocked with TBST (50mM Tris-HCl, pH 7.4,
150mM NaCl, and 0.1% Triton X-100; Sigma) containing
5% goat serum for 1 hour at RT. The coverslips were incu-
bated with rabbit polyclonal IgG (1/ml; Affinity Biore-
agents, Golden, CO) overnight at 4°C. On the following
day, after three washes with PBS for 5 minutes each, the
secondary antibody Alexa Fluor goat anti–rabbit IgG (Mo-
lecular Probes, Eugene, OR) were added to the coverslips
and incubated for 1 hour at RT. After washes with Tris-
buffered saline, nuclei were stained by adding Hoechst
33258 at a final concentration of 2g/ml for 1 minute. Cov-
erslips were mounted with FluoroMount (Southern Biotech,
Birmingham, AL) and kept in the dark at 4°C. Cell images
were captured with a fluorescence microscope (Nikon Eclipse
E800; Nikon, Du¨sseldorf, Germany) equipped with a Spot
RT digital camera (Diagnostic Instruments, Sterling Height,
MI).
IMMUNOBLOTTING. Cells were lysed in 1% sodium dode-
cyl sulfate buffer (pH 7.6, 20mM HEPES containing pro-
tease inhibitor; Roche, Mannheim, Germany), and lysates
were collected and sonicated for 12 seconds. Protein concen-
trations were determined using the Bio-Rad D
c
Protein assay
(Bio-Rad, Hercules, CA). Protein extracts and a biotinylated
molecular weight marker (Cell Signaling Technology, Bev-
erly, MA) were denaturated in Laemmli sample loading
buffer at 95°C, separated by 4 to 20% polyacrylamide gel
electrophoresis, and electrotransferred in transfer buffer to a
polyvinyl diflouride membrane (0.2m pore; Bio-Rad). The
membrane was treated with blocking solution (5% nonfat
dry milk in TBST) and incubated overnight at 4°C with
rabbit polyclonal ER-(1:200, 1g/ml; Santa Cruz Biotech-
nology, Santa Cruz, CA), rabbit polyclonal ER-(1:1,000,
1g/ml; Affinity BioReagents), rabbit polyclonal Akt
Gerstner et al: E2 Attenuates Brain Injury 563
(1:1,000), rabbit polyclonal phospho-Akt (1:1,000), poly-
clonal rabbit p44/42 mitogen-activated protein (MAP) ki-
nase (1:1,000), mouse monoclonal phospho-p44/42 MAP ki-
nase (1:1,000; all from Cell Signaling Technology, Danvers,
MA), or rabbit polyclonal Actin antibody (1:10,000; Sigma).
Secondary incubations were performed with horseradish per-
oxidase–linked anti–mouse (1:2,000; Bio-Rad) or anti–rabbit
(1:7,500; Amersham Pharmacia Biosciences, Bucks, United
Kingdom) antibodies. Bands were visualized using enhanced
chemiluminescence (PerkinElmer Life Sciences, Boston,
MA), and serial exposures were made to radiographic film
(Denville Scientific, Metuchen, NJ).
OXYGEN EXPOSURE. Cultures were kept in stage-specific
growth hormone–containing medium and transferred to a
humidified chamber filled with 80% O
2
plus 5% CO
2
plus
15% air at 37°C for different time periods. Control plates
were kept under 21% oxygen, indicated as normoxic, plus
5% CO
2
conditions at 37°C.
SURVIVAL ASSAY. After exposure to hyperoxia, cell culture
media were collected and centrifuged, and lactate dehydro-
genase activity was quantified in 50l samples of medium
supernatant, using a colorimetric cytotoxicity assay kit ac-
cording to the manufacturer’s instructions (Roche). Absor-
bance data were obtained using a 96-well plate reader (Mo-
lecular Devices, Sunnyvale, CA) with a 450nm filter, and
650nm as reference wavelength. Maximum lactate dehydro-
genase release was determined by cell lysis of one-well un-
treated cells with cell culture media containing 1% Triton
X-100.
ACTIVATED CASPASE-3 ASSAY. Activation of caspase-3 in
living cells was assessed by the caspase-3 activity assay
(Merck, Darmstadt, Germany) following the manufacturer’s
instructions. The assay utilizes a FITC-labeled monoclonal
antibody directed against the cleaved (activated) form of
caspase-3. The FITC label allows for direct detection of ac-
tivated caspase-3 in apoptotic cells by flow cytometry. Sam-
ples were analyzed by using the FL-1 channel of the flow
cytometer.
Animal Studies
All animal experiments were performed in accordance with
the guidelines of Humboldt University. Six-day-old Wistar
rat pups (BgVV, Berlin, Germany) were placed together with
their mothers in an oxygen chamber containing 80% oxygen
for defined time periods up to 24 hours. With beginning of
the exposure animals received a single injection of 600g/kg
E2 intraperitoneally, diluted in sterile water, volume 0.1ml/
10g (n 6). Control animals received an injection with ve-
hicle (n 6). Animals were decapitated at the end of expo-
sure at P7 and immediately removed for molecular studies.
For detection of myelin basic protein (MBP), animals were
killed 96 hours later at P11. Brains were immediately re-
moved for molecular studies. Tissue was either collected as
whole brain or was microdissected for corpus callosum and
adjacent white matter structures under the microscope, and
were subsequently snap-frozen in liquid nitrogen and stored
at 80°C until analysis.
IMMUNOBLOTTING OF BRAIN TISSUE. Snap-frozen tissue
(whole brain, white matter) was homogenized in RIPA buffer
(1% NP40, 0,5% sodium deoxycholate, 0,1% sodium dode-
cyl sulfate, 1mM EDTA, 1mM EGTA, 1mM Na
3
VO
4
,
20mM NaF, 0.5mM dithiothreitol (DTT), 1mM phenyl-
methyl sulfonyl fluoride, and protease inhibitor cocktail in
PBS pH 7.4). The homogenate was centrifuged at 1,050g
(4°C) for 10 minutes, and the microsomal fraction was sub-
sequently centrifuged at 17,000g(4°C) for 20 minutes.
Twenty micrograms of the resulting cytosolic protein extracts
were heat denaturated in Laemmli sample loading buffer,
separated by 10.5% sodium dodecyl sulfate polyacrylamide
gel electrophoresis, and electrotransferred onto a nitrocellu-
lose membrane. Nonspecific protein binding was prevented
by treating the membrane with 5% nonfat dry milk in Tris-
buffered saline/0.1% Tween 20 for 2 hours at RT. Thereaf-
ter, the membrane was incubated overnight at 4°C with pri-
mary antibody. For analysis of ER-/-rabbit polyclonal
antibodies (ER-; 1:500, ER-; 1:1,000; Affinity BioRe-
agents), of Fas a rabbit polyclonal antibody (1:250; Stress-
gen, Ann Arbor, MI), and of the mitogen-activated extracel-
lular kinase pathway, a rabbit polyclonal phosphorylation
state-independent extracellular signal-regulated kinases 1 and
2 (ERK1/2) antibody (1:1,000) and a mouse monoclo-
nal phospho-p44/42 ERK1/2 antibody (Thr202/Tyr204;
1:1,000) were used. The Akt pathway was analyzed by using
a rabbit polyclonal Akt antibody (1:1,000) and a rabbit poly-
clonal phospho-Akt antibody (Ser473; 1:1,000; all from Cell
Signaling). Secondary incubations were performed with
horseradish peroxidase–linked anti–mouse (1:2,000; Dako,
Glostrup, Denmark) or anti–rabbit (1:2,000; Amersham
Pharmacia Biosciences) antibody. Bands were visualized us-
ing enhanced chemiluminescence (Amersham Pharmacia
Biosciences), and serial exposures were made to radiographic
film (Hyperfilm enhanced chemiluminescence; Amersham
Pharmacia Biosciences). Densitometric analysis of the blots
was performed with an image analysis program (BioDocAna-
lyze; Whatman Biometra, Goettingen, Germany). For strip-
ping, membranes were incubated with stripping buffer
(100mM -mercaptoethanol, 2% sodium dodecyl sulfate,
62.5mM Tris-HCl, pH 6.7) at 50°C for 30 minutes, then
washed, blocked, and reprobed overnight at 4°C with mouse
anti–-actin monoclonal antibody (1:5,000; Sigma-Aldrich).
SEMIQUANTITATIVE REVERSE TRANSCRIPTASE POLYMER-
ASE CHAIN REACTION ON BRAIN TISSUE. Total cellular
RNA from snap-frozen whole brains was isolated by acidic
phenol/chloroform extraction
23
and DNase I treated
(Roche); 500ng RNA was reverse transcribed with Moloney
murine leukemia virus reverse transcriptase (Promega, Mad-
ison, WI) in 25l of reaction mixture. The resulting com-
plementary DNA (1l) was amplified by polymerase chain
reaction. Primers to amplify rat Fas (GenBank sequence
D26112) were 5-CCGACAACAACTGCTCAGA-3(sense
primer, positioned at nucleotide 174) and 5-GCACCTG-
CACTTGGTATTC-3(antisense primer, positioned at nu-
cleotide 430). The primers to amplify the internal standard
18S ribosomal RNA (GenBank sequence M11188) were 5-
AACGAGGATCCATTGGAG-3(sense primer, positioned
at nucleotide 596) and 5-ATGCCAGAGTCTCGTTCG-3
564 Annals of Neurology Vol 61 No 6 June 2007
(antisense primer, positioned at nucleotide 1409). Comple-
mentary DNA was amplified in 30 cycles, consisting of dena-
turing over 30 seconds at 94 °C, annealing over 45 seconds at
55°C, and primer extension over 45 seconds at 72°C. Ampli-
fied complementary DNA was subjected to 5% polyacryl-
amide gel electrophoresis, subsequent silver staining, and den-
sitometric analysis with the image analysis program
BioDocAnalyze (Whatman Biometra).
Statistical Analysis
Data were analyzed using GraphPad Prism version 4.00 for
Windows (GraphPad Software, San Diego, CA). Unless oth-
erwise indicated, the results shown are one experiment rep-
resentative of three to six separate experiments that were per-
formed. Either Student’s ttest, one-way analysis of variance,
or two-way analysis of variance with the Tukey–Kramer post
hoc analysis for multiple comparisons were performed for
statistical analyses. Statistical significance was determined at
p0.05.
Results
Estrogen Receptor-
and -
Are Expressed in
Oligodendroglial Cells during Development
To determine the expression of ER-and -during
oligodendroglial cell development, we assessed protein
levels of both receptors in developing primary OL cul-
tures (Fig 1A). ER-and -were preferentially ex-
pressed in immature OLs and showed a downregula-
tion in mature OLs. Furthermore, the intracellular
distribution of ER-was examined in pre-OLs by im-
munocytochemistry (see Fig 1B). ER-was predomi-
nantly located in the nucleus, with diffuse cytoplasmic
staining.
Time Dependency of Hyperoxia-Induced Toxicity to
Primary Oligodendrocytes
To estimate the time course of oxygen toxicity, pri-
mary pre-OLs were incubated with 80% oxygen for 0,
6, 12, and 24 hours. Cell viability was assessed by lac-
tate dehydrogenase release. Survival of developing OLs
decreased with time of exposure (Fig 2). After 12 hours
of incubation, cell survival was reduced to 25%, and
after 24 hours almost no viable cells were detected.
Control cells were kept in 21% oxygen for 0 to 24
hours and displayed a constant amount (8–10%) of
cell death for each time period (data not shown).
17
-Estradiol Protects against Hyperoxia-Mediated
Cell Death in Developing Oligodendrocytes
To investigate whether E2 treatment can reduce
hyperoxia-mediated cell death in OLs, we administered
E2 or vehicle to primary pre-OL cultures at 0, 2, 4, 6,
12, 24, and 48 hours before 12 hours exposure to 80%
oxygen. To address whether the neuroprotective effect
of E2 is dose dependent, we incubated pre-OLs with
80% oxygen without (Fig 3A), with 1M E2 (see Fig
3B), or with 1nM E2 (see Fig 3C) 12 hours before
oxygen exposure. Representative transmission light-
phase contrast photomicrographs of primary OL cell
cultures showed that after 12 hours of hyperoxia, de-
veloping OLs showed signs of cell death including
plasma membrane blebbing and nuclear condensation.
Cells that were pretreated with E2 display less cell
death that was dose dependent with a median effective
concentration (EC
50
)of4.710
10
M (see Fig 3D).
Significant protection with 1M E2 was found, if the
drug was given to the cell culture at least 4 hours be-
fore exposure to 12 hours of 80% oxygen (see Fig 3E).
Fig 1. (A) Western blot analysis of estrogen receptors (ER)-
and -
expression in premyelinating (O4
) and mature
(MBP
) oligodendrocytes showing a specific band at 66kDa
for the ER-
and a specific band at 55kDa for the ER-
whose intensities were found to be decreased in mature OLs in
comparison with the
-actin band (42kDa). Cell lysates were
immunoblotted with antibodies against ER-
, ER-
, and
-actin, as indicated. (B) Immunocytochemistry of ER-
in
immature oligodendrocytes (O4
). ER-
(green) was mostly
nuclear (Hoechst 33258, blue), but also showed a diffuse dis-
tribution in the cytoplasm. Scale bar 10
m. MBP mye-
lin basic protein.
Fig 2. Hyperoxia causes cell death in developing oligodendro-
cytes. Primary oligodendrocyte cell viability, as measured by lac-
tate dehydrogenase release, after 0, 6, 12, and 24 hours incuba-
tion with 80% oxygen (means standard error of the mean of
3 independent experiments). *p0.05; ***p0.001, one-
way analysis of variance with Tukey’s multiple-comparison tests.
Gerstner et al: E2 Attenuates Brain Injury 565
Fas Death Receptor Is Involved in Apoptotic Cell
Death in Developing Oligodendrocytes
Fas is a key molecule serving in most cases as a death
signal within the apoptotic machinery.
24
Increase in
expression of Fas after 80% oxygen exposure in the ol-
igodendroglia cell line was determined at different time
points after insult. Immunoblotting demonstrated an
increase of Fas at 6 and 12 hours after 80% oxygen
exposure in the oligodendroglia cell line. Pretreatment
with 100nM E2 attenuated this upregulation, signifi-
cant after 6 hours of oxygen exposure (Fig 4A). Previ-
ously, we reported on the distribution pattern and type
of neurodegeneration induced by hyperoxia in the de-
veloping brain of Wistar rats. Degenerating cells were
determined in the frontal, parietal, cingulate, and ret-
rosplenial cortex, caudate nucleus, nucleus accumbens,
corpus callosum and adjacent white matter, thalamus,
hippocampal dentate gyrus, subiculum, and hypothal-
amus.
8
Application of E2 intraperitoneal substantially
reduced cumulative cell death scores in the P6 rat brain
after exposure to 80% oxygen for 24 hours.
25
In this study, an increase in expression of Fas in the
brains of 6-day-old rats after 80% oxygen exposure was
determined at defined time points after insult using re-
verse transcriptase polymerase chain reaction and West-
ern blotting. Reverse transcriptase polymerase chain re-
action and immunoblotting was performed in samples
from whole brain and white matter preparations. Poly-
acrylamide gels (Fas in reference to 18S ribosomal
RNA) and blots (Fas in reference to -actin band)
Fig 3. (A–C) Neuroprotective effect of 17
-estradiol (E2) against hyperoxia-mediated cell death. Representative transmission light-
phase contrast photomicrographs of primary oligodendrocyte cell cultures after 12 hours incubation with either 21% (data not
shown) or 80% oxygen. After incubation with 80% oxygen, immature oligodendrocytes (OLs) without (A), with 1
M E2 (B), and
with 1nM E2 (C) treatment. Hyperoxia-exposed immature OLs show signs of cell death including plasma membrane blebbing and
nuclear condensation. Cells that were pretreated with E2 display less cell death that is dose dependent. (D) Dose-dependent effect of
E2. Treatment of primary OL cell cultures with E2 at different concentrations (10
6
,10
7
,10
8
,10
9
,10
10
M, 12 hours be-
fore treatment), followed by exposure to 80% oxygen for 12 hours. Cell toxicity was measured by lactate dehydrogenase (LDH) re-
lease (optical density ratio). The protective effect of E2 dose dependent with an median effective concentration (EC
50
)of4.7
10
10
M. Means standard error of the mean (SEM) of three independent experiments. **p0.01, ***p0.001 in one-way
analysis of variance with Tukey’s multiple-comparison tests. (E) Pretreatment of primary OL cell cultures with E2 0, 4, 6, 12, 24,
and 48 hours before hyperoxia. Percentage of survival was measured by LDH release. The protective effect of E2 was significant
after pretreatment of at least 4 hours. Means SEM of three independent experiments. ***p0.001; *p0.05, one-way anal-
ysis of variance with Tukey’s multiple-comparison tests.
566 Annals of Neurology Vol 61 No 6 June 2007
were subjected to densitometric analysis, and density
ratios were statistically analyzed. Hyperoxia triggered
an increase of Fas messenger RNA (see Fig 4B) and Fas
protein (see Figs 4C, D) after 12 and 24 hours in the
P6-day-old rat brain. Treatment of P6-day-old rats
with E2 (600g/kg intraperitoneally) before hyperoxia
Fig 4. (A) Effect of 17
-estradiol (E2) on apoptosis signaling proteins in immature oligodendrocytes (OLN-93) exposed to hyper-
oxia. Immunoblot densitometric quantification of protein levels for Fas shows increased Fas-expression after 6 and 12 hours of hy-
peroxia. ***p0.001, **p0.01, comparison between normoxia (0 hours) or oxygen exposure for 2, 6, 12, or 24 hours (analy-
sis of variance). Treatment with estrogen (100nM) eliminates Fas-upregulation. Blot is representative of a series of blots of three
independent experiments. Graphs represent means standard error of means (SEMs) of the density ratios of Fas to
-actin. #p
0.05, comparison between vehicle and E2-treated cells, Student’s ttest. (B) Fas messenger RNA (mRNA) expression in P7-day-old
whole rat brains 12 or 24 hours after 80% oxygen exposure and in P6-day-old whole rat brains treated with E2 before hyperoxia.
There was an increase in mRNA levels for Fas 12 and 24 hours after hyperoxia. Treatment of P6-day-old rats with E2 before hy-
peroxia exposure attenuated Fas upregulation in comparison with Fas-mRNA levels of untreated littermates. The results of densito-
metric analysis of the gels are presented in reference to 18S ribosomal RNA (rRNA). Data represent the ratio (%) of the density of
the Fas band to the 18S rRNA band SEM (n 4). *p0.05, comparison between P7-day-old whole rat brains 12 or 24
hours after 80% oxygen exposure and P7-day-old whole rat brains treated with E2 before hyperoxia (analysis of variance). (C, D)
Hyperoxia triggered an increase of Fas protein after 12 and 24 hours in lysates from P7-day-old whole rat brain (WB) and white
matter preparation (WM). Treatment of P6-day-old rats with E2 (600
g/kg intraperitoneally) before hyperoxia exposure attenuated
Fas upregulation in comparison with Fas levels of untreated littermates. Blots are representative of a series of four blots. Densitomet-
ric quantification of the blots is presented. Columns represent means SEMs of the density ratios of Fas to
-actin (n 4 per
group). ***p0.001, comparison between vehicle and E2, Student’s ttest.
Gerstner et al: E2 Attenuates Brain Injury 567
exposure attenuated Fas upregulation in comparison
with Fas levels of untreated littermates.
Hyperoxia Induces Activation of Caspase-3 in the
Oligodendroglia Cell Line OLN-93 That Is
Attenuated after Pretreatment with 17
-Estradiol
Caspase-3 has been shown to play a key role in medi-
ating the effector stage of apoptosis by initiating the
process of DNA fragmentation.
26
The activation of the
effector caspase-3 was assessed by intracellular binding
of a FITC-labeled monoclonal antibody specific to the
activated (cleaved) form of the enzyme using flow cy-
tometry. As described previously, after 24 hours of ox-
ygen incubation, OLN-93 cell viability is reduced to
80%, compared with 10 to 20% in primary OLs, and
only after 96 hours oxygen exposure, almost no
OLN-93 cells are viable.
10
Primary OLs are much
more susceptible toward oxygen exposure (see Fig 2).
The percentage of OLs displaying activated caspase-3
was found to increase up to twofold after a 24-hour
incubation period with 80% oxygen (Fig 5). Pretreat-
ment with 100nM E2 significantly reduced caspase-3
activation by approximately 30%.
Hyperoxia Reduces Levels of Phosphorylated
Extracellular Signal–Regulated Kinase 1/2 and Akt
in Primary Oligodendroglia Cultures and in the
Brains of 7-Day-Old Rats That Is Reversed
by 17
-Estradiol
The kinases ERK1/2 and Akt (protein kinase B) are
members of two important pathways that control neu-
ronal cell survival, the MAP kinase and the phosphati-
dylinositol 3 (PI3) kinase pathways. Activated ERK1/2
recently was shown to phosphorylate and inhibit
caspase-9, a key caspase in activation of the intrinsic ap-
optotic pathway, which is initiated by release of cyto-
chrome cinto the cytoplasm.
27
Moreover, it has been
shown that activated Akt phosphorylates Bad, a pro-
apoptotic molecule, and causes its inactivation. This
leads to liberation and activation of the antiapoptotic
molecule Bcl-2.
28
To investigate the impact of treatment
with E2 on levels of phosphorylated (active) ERK1/2
and Akt, we performed Western blot analysis on protein
samples obtained from primary oligodendroglia cultures
(Figs 6A, B) and white matter preparations (see Figs 6C,
D) from P6 rats subjected to 80% oxygen for various
time points or room air. This analysis indicated that ex-
posure to 80% oxygen decreased levels of the active,
phosphorylated isoforms of the ERK1/2 (pERK1/2) (see
Figs 6B, D) and of the serine-threonine kinase Akt
(pAkt) (see Figs 6A, C), which both mediate intracellu-
lar signaling after plasma membrane-associated tyrosine
receptor kinase autophosphorylation by growth fac-
tors.
29
E2 ameliorated hyperoxia-induced decreases of
the phosphorylated active forms of pERK1/2 and pAkt
in primary developing OLs and white matter prepara-
tion of the P6 rat brain.
TREATMENT OF 17-ESTRADIOL ATTENUATES WHITE
MATTER INJURY IN VIVO. Our group recently found
that oxygen-induced cell death in vivo is age depen-
dent.
8
Rats at postnatal ages P0, P3, P7, P14, and P20
were subjected to 80% oxygen over a 24-hour period.
After oxygen exposure, degenerating cells were quanti-
fied in sections stained by the DeOlmos cupric silver
method inter alia in the corpus callosum and adjacent
white matter.
8
Here we investigated whether oxygen
influences the developing process of OLs in vivo by
subjecting P6 rat pups to 80% oxygen over a 24-hour
period and killing the animals at P11. At the beginning
of the hyperoxia exposure, rat pups were treated intra-
peritoneally with E2 (600g/kg) or vehicle (2-
hydroxypropyl--cyclodextrin). The injury was assessed
by two blinded observers for degree of MBP loss in the
cerebral white matter (Fig 7A–D). There was an atten-
uation of lesion severity in rats treated with E2, as
compared with vehicle-treated control animals (see Figs
7C, D).
Discussion
This study demonstrates that E2 treatment is protec-
tive in hyperoxia-induced white matter injury in the
developing rat brain. E2 acts in a neuroprotective fash-
ion by downregulation of Fas death receptor, inhibi-
tion of caspase-3 activation, restoration of survival sig-
nals transmitted by MAP kinases and protein kinase B,
and attenuation of MBP loss in developing OLs in
vivo.
Recently, we reported that exposure of the brain to
high concentrations of oxygen cause apoptotic cell
Fig 5. 17
-Estradiol (E2) decreases activation of caspase-3.
Activation of the effector caspase-3 was assessed by intracellular
binding of a fluorescein isothiocyanate–labeled monoclonal
antibody specific for the activated (cleaved) form of the en-
zyme, using flow cytometry. OLN-93 cells were incubated un-
der hyperoxic (80% oxygen) conditions for 24 hours.
Means standard error of the mean of three independent
experiments. **p0.01 in Student’s ttest.
568 Annals of Neurology Vol 61 No 6 June 2007
death during a specific period of development in vitro
and in vivo. Cultured pre-OLs (O4
O1
MBP
) and
immature OLs (O4
O1
MBP
), derived from the
OLN-93 cell line, display susceptibility toward hyper-
oxia, whereas mature OLs (MBP
) remain viable.
30
Moreover, we reported that hyperoxia induces wide-
spread cell death in brains of 3- to 6-day-old rats and
mice, both in gray and white matter regions.
8
Vulner-
ability to oxygen-induced cell death was age dependent
with a maximum during the first week of life in both
OL cell cultures and animals.
Several studies have highlighted that female sex hor-
mones represent potential neuroprotective agents
against damage produced by acute and chronic injuries
in the adult brain. Estrogens have been shown to pro-
mote survival and differentiation of several neuronal
populations maintained in culture
31
and to reduce cell
death associated with excitotoxicity,
15
oxidative
stress,
32
or exposure to -amyloid.
33
The neuroprotec-
tive effects of estrogen have been widely documented
in animal models of neurological disorders, such as
Parkinson’s
34
and Alzheimer’s diseases,
35
as well as ce-
rebral ischemia.
14
Both ER-and -are found in various regions of
the human and rodent brain, including the hypothala-
mus, hippocampus, cerebral cortex, midbrain, brain-
Fig 6. (A, B) 17
-Estradiol (E2) reversed hyperoxia-induced reduction of phosphorylated isoforms of extracellular signal–regulated ki-
nase 1 and 2 (pERK1/2) and pAkt levels in primary oligodendrocytes (OLs). pAkt (A) and pERK1/2 (B) levels were reduced in O4
OLs after hyperoxia exposure. Total ERK1/2 and Akt levels remained unaltered. Administration of E2 increased pERK1/2 and pAkt
levels compared with cells that were treated with vehicle. Blots are representative of a series of three blots for each antibody and each
treatment condition. Densitometric quantification of the blots is presented. Columns represent means standard error of the means
(SEMs) of the density ratios of the protein of interest to
-actin (n 4 per group). Treatment with E2 increased levels of pERK1/2
and pAkt in comparison with vehicle-treated oligodendrocytes and leaves levels of ERK1/2 and Akt unaltered. ***p0.001; **p
0.01; *p0.05, comparison between vehicle and E2 (B), Student’s ttest. (C, D) E2 reversed hyperoxia-induced reduction of phos-
phorylated isoform of pAkt (C) and pERK1/2 (D) levels in the white matter of P7-day-old rats. Administration of E2 (600
g/kg in-
traperitoneally) increased pAkt and pERK1/2 levels significantly and leaves levels of Akt and ERK1/2 unaltered. Blots are representative
of a series of six blots for each antibody. Densitometric quantification of the blots is presented. Columns represent means SEMs of
the density ratios of the protein of interest to
-actin (n 4 6 per group). (C) ***p0.001; **p0.01; *p0.05, comparison
between vehicle and E2, Student’s ttest. (D) ***p0.001; **p0.01, comparison between vehicle and E2; ##p0.01, compar-
ison between control and hyperoxia treated P7-day-old rat pups, Student’s ttest.
Gerstner et al: E2 Attenuates Brain Injury 569
stem, and forebrain, and ER-mediated effects are
thought to provide neuroprotection.
11,36
These studies
defined the neuroanatomic localization of each recep-
tor, but did not specify cell type. Here, we report that
ER-and -are expressed in oligodendroglial cells.
Possible mechanisms underlying estrogen’s protective
effects include the activation of nuclear, cytoplasmic,
and membrane-localized ER. Interestingly, our data
demonstrate that ER-protein expression displays a
downregulation during the maturation process of oli-
godendroglial cells, suggesting an important role of
ER-during brain development and in models of neo-
natal brain injury. In an ovariectomized/ischemia adult
mouse model of stroke, estradiol treatment protects
wild-type and ER-–null mice from brain injury,
whereas this protection is abolished in ER-–null ani-
mals, suggesting a crucial role for ERin providing
neuroprotection.
37
However, another study in which
stroke was induced by reversible middle cerebral artery
occlusion found no increase in tissue damage in ER-
–null mice, suggesting that non–ER-mediated path-
ways, involving antioxidant effects, interaction with
membrane binding sites, and modulation of neuro-
transmitter systems, may be involved.
15,32,38
Therefore,
protection by estrogen may involve both ER-dependent
classic and nonclassic genomic responses, and ER-
independent mechanisms. Thus, further studies are
warranted to better characterize the modes of action of
estrogen in the brain.
Encouraged by the aforementioned work of others in
neuronal cultures and adult animal models of brain in-
jury, we set out to study the effect of estradiol in the
neonatal brain, focusing on the white matter. Espe-
cially during the last trimester of pregnancy, migration
Fig 7. (A, B) Normal myelin basic protein (MBP) expression in P11 rat pup that was kept under normoxic conditions. (C, D)
Bilateral loss of MBP is seen in the P11 pup in the white matter tract after hyperoxia for 24 hours at P6. (E, F) Protective effect
of 17
-estradiol in vivo, demonstrating attenuation of MBP injury at P11 with systemic 17
-estradiol treatment during hyperoxia
exposure (n 3 per group).
570 Annals of Neurology Vol 61 No 6 June 2007
and differentiation of OLs take place.
39
Late OL pro-
genitors (O4
O1
MBP
) and immature OLs
(O4
O1
MBP
) are the predominant OL stage in
human cerebral white matter during the peak incidence
of PVL.
40
When the umbilical cord is clamped, high
placental estrogen supply is no longer available for the
premature neonate. This may in itself lead to disadvan-
tages or a greater risk for development of PVL and
later motor and cognitive deficits. However, based on
Nilsen and colleagues’ data,
41
proapoptotic effects of
E2 in the immature brain cannot be excluded.
One potential mechanism for the protective effect of
E2 includes changes in gene expression in the develop-
ing brain and in oligodendroglial cultures; that is, ac-
tivation of cell survival–promoting signaling pathways,
such as the mitogen-activated extracellular kinase-
ERK1/2 and the PI3 kinase-Akt pathways. As de-
scribed previously, hyperoxia reduced levels of the ac-
tive forms of ERK1/2 and Akt in a time-dependent
fashion in thalamus, striatum, and cortical structures.
8
ERK1/2 and Akt are key players in these two path-
ways, which are activated by tyrosine kinase receptors
on binding of growth factors to their receptors. These
changes reflect impairment of survival promoting sig-
nals
42
and imbalance between neuroprotective and
neurodestructive mechanisms in the brain, which dur-
ing the physiological, developmental elimination of
brain cells will likely promote apoptotic death. Here
we demonstrate that the hyperoxia-mediated down-
regulation of the phosphorylated forms of ERK and
Akt occurs also in the white matter of neonatal rats
and in oligodendroglial cells, indicating an important
role of these pathways in the pathophysiology of white
matter injury induced by hyperoxia.
Activation of ERK1/2, and activation of Akt, can
ameliorate hyperoxia-induced apoptotic cell death in
lungs and retina.
43,44
Other groups have shown that
the PI3 kinase cascade is involved in the neuroprotec-
tive mechanism stimulated by estrogen.
25,45
The con-
cept that attenuation of pAkt loss may contribute to
the protective effect of E2 is well-founded; however,
previous data on pERK in neonatal and adult brain
injury indicate that this MAP kinase has both proapop-
totic and antiapoptotic actions.
46
This study shows
that E2 counteracts the inactivation of ERK1/2 and
Akt pathways, and therefore protects against apoptotic
cell death. A previous in vivo study supports the fact
that oxygen causes cell death by inactivation of survival
signaling proteins Ras, ERK-1/2, and protein kinase B
(Akt). Furthermore, synRas-transgenic mice overex-
pressing constitutively activated Ras and phosphory-
lated kinases ERK-1/2 in the brain were protected
against oxygen neurotoxicity.
8
Another pathway that accounts for the antiapoptotic
action of E2 is its influence on the expression level of
the proapoptotic molecule Fas. Here we present evi-
dence that hyperoxia leads to increased expression of
Fas in the P7 rat brain and in premyelinating OLs.
Treatment of P6 rats with E2 before hyperoxia expo-
sure attenuated Fas-messenger RNA upregulation in
comparison with Fas-messenger RNA levels of un-
treated littermates and decreased Fas protein expression
in the oligodendroglial cell line. Others have shown
that female steroid hormones can influence the apopto-
tic cascade at different stages. Injury models relying on
apoptotic signaling mechanisms have demonstrated
neuroprotection via estrogen inhibition of proapoptotic
factors, including caspases,
47,48
cytochrome c,
49
and ac-
tivation of antiapoptotic gene transcription, such as
Bcl-2 and Bcl-XL.
50,51
Furthermore, E2 reduces the
proapoptotic calcium influx into neuronal cells.
16
Moreover, E2 has antioxidative properties that can lead
to the reduction of free radicals.
17
Bittigau and co-
workers
52
showed in a P7 model of antiepileptic drug–
induced brain injury that injection of E2 significantly
reduces neurodegeneration by activation of antiapop-
totic signaling pathways involving phosphatidylinositol
3-Akt and MAP kinases.
In this study, we demonstrated the protective impact
of E2 on the developing white matter. Our results raise
the possibility that specific hormonal replacement
treatment for premature infants may improve neuro-
logical outcome. These results raise the interesting hy-
pothesis that E2 might be suitable as a preventive agent
in premature infants and sick term infants, who need
to be exposed to oxygen for the purposes of treatment.
However, determination of an exact time window is
highly warranted as a focus of further studies. E2 re-
placement therapy in extremely low-birth-weight in-
fants has been introduced in some centers with the
goal to improve bone mineralization, and no adverse
side effects have been observed so far.
53
Therefore, we
suggest that maintaining placental E2 plasma levels
may be effective to protect neonates from brain injury.
More data are urgently needed concerning the safety
and feasibility of estrogen supplementation in the neo-
natal period.
This work was supported by the Ernst Schering Research Founda-
tion, the German Federal Department of Education and Research
(BMBF; 01 ZZ 0101), the Sonnenfeld-Stiftung, the European
Commission (Sixth Framework Program, contract no LSHM-CT-
2006-036534), and the NIH (NS28475, HD18655, PO1NS38475,
J.J.V., P.A.R.).
We are grateful to Prof Dr Richter-Landsberg, who kindly provided
us with OLN-93 cells.
Gerstner et al: E2 Attenuates Brain Injury 571
References
1. Hack M, Flannery DJ, Schluchter M, et al. Outcomes in young
adulthood for very-low-birth-weight infants. N Engl J Med
2002;346:149–157.
2. Ment LR, Vohr B, Allan W, et al. Change in cognitive function
over time in very low-birth-weight infants. JAMA 2003;289:
705–711.
3. Marlow N, Wolke D, Bracewell MA, Samara M. Neurologic
and developmental disability at six years of age after extremely
preterm birth. N Engl J Med 2005;352:9–19.
4. Ajayi-Obe M, Saeed N, Cowan FM, et al. Reduced develop-
ment of cerebral cortex in extremely preterm infants. Lancet
2000;356:1162–1163.
5. Nosarti C, Al-Asady MH, Frangou S, et al. Adolescents who
were born very preterm have decreased brain volumes. Brain
2002;125:1616–1623.
6. Dobbing J, Sands J. Comparative aspects of the brain growth
spurt. Early Hum Dev 1979;3:79 83.
7. Volpe JJ. Perinatal brain injury: from pathogenesis to neuro-
protection. Ment Retard Dev Disabil Res Rev 2001;7:56 64.
8. Felderhoff-Mueser U, Bittigau P, Sifringer M, et al. Oxygen
causes cell death in the developing brain. Neurobiol Dis 2004;
17:273–282.
9. Felderhoff-Mueser U, Sifringer M, Polley O, et al. Caspase-1-
processed interleukins in hyperoxia-induced cell death in the
developing brain. Ann Neurol 2005;57:50–59.
10. Gerstner B, Bu¨hrer C, Rheinla¨nder C, et al. Maturation-
dependent oligodendrocyte apoptosis caused by hyperoxia.
J Neurosci Res 2006;84:306–315.
11. Shughrue PJ, Lane MV, Merchenthaler I. Comparative distri-
bution of estrogen receptor-alpha and -beta mRNA in the rat
central nervous system. J Comp Neurol 1997;388:507–525.
12. Wang L, Andersson S, Warner M, Gustafsson JA. Estrogen re-
ceptor (ER)beta knockout mice reveal a role for ERbeta in mi-
gration of cortical neurons in the developing brain. Proc Natl
Acad SciUSA2003;100:703–708.
13. Mitra SW, Hoskin E, Yudkovitz J, et al. Immunolocalization of
estrogen receptor beta in the mouse brain: comparison with es-
trogen receptor alpha. Endocrinology 2003;144:2055–2067.
14. Merchenthaler I, Dellovade TL, Shughrue PJ. Neuroprotection
by estrogen in animal models of global and focal ischemia. Ann
NY Acad Sci 2003;1007:89–100.
15. Weaver CE Jr, Park-Chung M, Gibbs TT, Farb DH. 17beta-
Estradiol protects against NMDA-induced excitotoxicity by di-
rect inhibition of NMDA receptors. Brain Res 1997;761:
338–341.
16. Mermelstein PG, Becker JB, Surmeier DJ. Estradiol reduces
calcium currents in rat neostriatal neurons via a membrane re-
ceptor. J Neurosci 1996;16:595–604.
17. Behl C. Estrogen can protect neurons: modes of action. J Ste-
roid Biochem Mol Biol 2002;83:195–197.
18. Brannvall K, Korhonen L, Lindholm D. Estrogen-receptor-
dependent regulation of neural stem cell proliferation and dif-
ferentiation. Mol Cell Neurosci 2002;21:512–520.
19. Tulchinsky D, Hobel CJ, Yeager E, Marshall JR. Plasma estrone,
estradiol, estriol, progesterone, and 17-hydroxyprogesterone in
human pregnancy. I. Normal pregnancy. Am J Obstet Gynecol
1972;112:1095–1100.
20. Richter-Landsberg C, Heinrich M. OLN-93: a new permanent
oligodendroglia cell line derived from primary rat brain glial
cultures. J Neurosci Res 1996;45:161–173.
21. Rosenberg PA, Dai W, Gan XD, et al. Mature myelin basic
protein-expressing oligodendrocytes are insensitive to kainate
toxicity. J Neurosci Res 2003;71:237–245.
22. Li J, Lin JC, Wang H, et al. Novel role of vitamin K in pre-
venting oxidative injury to developing oligodendrocytes and
neurons. J Neurosci 2003;23:5816–5826.
23. Chomczynski P, Sacchi N. Single-step method of RNA isola-
tion by acid guanidinium thiocyanate-phenol-chloroform ex-
traction. Anal Biochem 1987;162:156–159.
24. Fas SC, Fritzsching B, Suri-Payer E, Krammer PH. Death re-
ceptor signaling and its function in the immune system. Curr
Dir Autoimmun 2006;9:1–17.
25. Asimiadou S, Bittigau P, Felderhoff-Mueser U, et al. Protection
with estradiol in developmental models of apoptotic neurode-
generation. Ann Neurol 2005;58:266–276.
26. Lavrik IN, Golks A, Krammer PH. Caspases: pharmacological
manipulation of cell death. J Clin Invest 2005;115:2665–2672.
27. Allan LA, Morrice N, Brady S, et al. Inhibition of caspase-9
through phosphorylation at Thr 125 by ERK MAPK. Nat Cell
Biol 2003;5:647–654.
28. Osterhout DJ, Marin-Husstege M, Abano P, Casaccia-Bonnefil
P. Molecular mechanisms of enhanced susceptibility to apopto-
sis in differentiating oligodendrocytes. J Neurosci Res 2002;69:
24–29.
29. Lee R, Kermani P, Teng KK, Hempstead BL. Regulation of cell
survival by secreted proneurotrophins. Science 2001;294:
1945–1948.
30. Gerstner B, Buhrer C, Rheinlander C, et al. Maturation-
dependent oligodendrocyte apoptosis caused by hyperoxia.
J Neurosci Res 2006;84:306–315.
31. Lustig RH. Sex hormone modulation of neural development in
vitro. Horm Behav 1994;28:383–395.
32. Sawada H, Ibi M, Kihara T, et al. Estradiol protects mesence-
phalic dopaminergic neurons from oxidative stress-induced neu-
ronal death. J Neurosci Res 1998;54:707–719.
33. Zhao L, Brinton RD. Select estrogens within the complex for-
mulation of conjugated equine estrogens (Premarin) are protec-
tive against neurodegenerative insults: implications for a com-
position of estrogen therapy to promote neuronal function and
prevent Alzheimer’s disease. BMC Neurosci 2006;7:24.
34. Sawada H, Ibi M, Kihara T, et al. Estradiol protects dopami-
nergic neurons in a MPP
Parkinson’s disease model. Neuro-
pharmacology 2002;42:1056–1064.
35. Baum LW. Sex, hormones, and Alzheimer’s disease. J Gerontol
A Biol Sci Med Sci 2005;60:736–743.
36. Laflamme N, Nappi RE, Drolet G, et al. Expression and neu-
ropeptidergic characterization of estrogen receptors (ERalpha
and ERbeta) throughout the rat brain: anatomical evidence of
distinct roles of each subtype. J Neurobiol 1998;36:357–378.
37. Dubal DB, Zhu H, Yu J, et al. Estrogen receptor alpha, not
beta, is a critical link in estradiol-mediated protection against
brain injury. Proc Natl Acad Sci USA 2001;98:1952–1957.
38. Sampei K, Goto S, Alkayed NJ, et al. Stroke in estrogen
receptor-alpha-deficient mice. Stroke 2000;31:738–744.
39. Levison SW, Goldman JE. Both oligodendrocytes and astro-
cytes develop from progenitors in the subventricular zone of
postnatal rat forebrain. Neuron 1993;10:201–212.
40. Back SA, Luo NL, Borenstein NS, et al. Late oligodendrocyte
progenitors coincide with the developmental window of vulner-
ability for human perinatal white matter injury. J Neurosci
2001;21:1302–1312.
41. Nilsen J, Mor G, Naftolin F. Estrogen-regulated developmental
neuronal apoptosis is determined by estrogen receptor subtype
and the Fas/Fas ligand system. J Neurobiol 2000;43:64–78.
42. Heumann R. Neurotrophin signalling. Curr Opin Neurobiol
1994;4:668 679.
43. Harms C, Lautenschlager M, Bergk A, et al. Differential mecha-
nisms of neuroprotection by 17 beta-estradiol in apoptotic versus
necrotic neurodegeneration. J Neurosci 2001;21:2600–2609.
572 Annals of Neurology Vol 61 No 6 June 2007
44. Buckley S, Driscoll B, Barsky L, et al. ERK activation protects
against DNA damage and apoptosis in hyperoxic rat AEC2.
Am J Physiol 1999;277:L159–L166.
45. Honda K, Sawada H, Kihara T, et al. Phosphatidylinositol
3-kinase mediates neuroprotection by estrogen in cultured cor-
tical neurons. J Neurosci Res 2000;60:321–327.
46. Zhuang S, Schnellmann RG. A death-promoting role for extra-
cellular signal-regulated kinase. J Pharmacol Exp Ther 2006;
319:991–997.
47. Jover T, Tanaka H, Calderone A, et al. Estrogen protects
against global ischemia-induced neuronal death and prevents
activation of apoptotic signaling cascades in the hippocampal
CA1. J Neurosci 2002;22:2115–2124.
48. Kajta M, Trotter A, Lason W, Beyer C. Impact of 17beta-
estradiol on cytokine-mediated apoptotic effects in primary hip-
pocampal and neocortical cell cultures. Brain Res 2006;1116:
64–74.
49. Bagetta G, Chiappetta O, Amantea D, et al. Estradiol reduces
cytochrome c translocation and minimizes hippocampal damage
caused by transient global ischemia in rat. Neurosci Lett 2004;
368:87–91.
50. Zhao L, Wu TW, Brinton RD. Estrogen receptor subtypes al-
pha and beta contribute to neuroprotection and increased Bcl-2
expression in primary hippocampal neurons. Brain Res 2004;
1010:22–34.
51. Pike CJ. Estrogen modulates neuronal Bcl-xL expression and
beta-amyloid-induced apoptosis: relevance to Alzheimer’s dis-
ease. J Neurochem 1999;72:1552–1563.
52. Bittigau P, Sifringer M, Genz K, et al. Antiepileptic drugs and
apoptotic neurodegeneration in the developing brain. Proc Natl
Acad SciUSA2002;99:15089–15094.
53. Trotter A, Bokelmann B, Sorgo W, et al. Follow-up examina-
tion at the age of 15 months of extremely preterm infants after
postnatal estradiol and progesterone replacement. J Clin Endo-
crinol Metab 2001;86:601–603.
Gerstner et al: E2 Attenuates Brain Injury 573
... The fetus is constantly exposed to the changing hormone levels during its crucial stages of development. Estrogen is considered to be important for normal brain development, and several experimental studies have shown that it can act as a potential neuroprotective agent against acute and chronic injuries in the brain [8][9][10]. ...
... However, preterm-born infants experience a drastic drop in estradiol (E2) and progesterone levels within 24 hours along with a dramatic increase of the oxygen tissue tension at a much earlier phase of brain development as compared to infants born at term [9]. This led to the idea of supplementation of these hormones for their neuroprotective properties [11]. ...
... This was achieved at the 80% O 2 concentration. Moreover, 80% O 2 has been widely used to induce cell death/disrupt myelin formation in experimental white matter disease, and the effects are well characterized [9,37,39]. Furthermore, we selected 24 hours as an appropriate incubation time for the treatment with hormones, as within this time period, we could observe the effects of the ...
Article
Full-text available
Cerebral oxygenation disturbances contribute to the pathogenesis of brain lesions in preterm infants with white matter damage. These children are at risk of developing long-term neurodevelopmental disabilities. Preterm birth is associated with sudden hormonal changes along with an untimely increase in oxygen tissue tension. There is a persistent high postnatal production of fetal zone steroids (FZS), which serve in the fetoplacental unit as precursors for placental estrogen synthesis during pregnancy. The role of FZS in events associated with oxygenation differences and their impact on the developing white matter is not well understood. Therefore, we investigated the effect of hyperoxia (80% O2) and subsequent administration of FZS on the protein composition and migration capabilities of immature oligodendrocytes using the OLN93 (rat-derived OPC) cell line as an experimental model. We tested the effect of the FZS, dehydroepiandrosterone (DHEA), 16α-OH-DHEA, and adiol (5-androstene-3β, 17β-diol). After 24-hour exposure to hyperoxia, we monitored the changes in the proteome profile following treatment and observed significant alterations in pathways regulating cytoskeletal remodelling, cell migration, and cell survival. Additionally, hyperoxia leads to impaired migration of the OLN93 cells in culture. Administration of the FZS showed positive effects on the migration process under normoxic conditions in general. However, under hyperoxic conditions, the trend was less prominent. The observed effects could be related to changes in levels of cofilin/LIMK pathway-associated proteins. Adiol had a negative effect when administered together with estradiol, and the proteomic data reveal the activation of ephrin receptor signalling that might be responsible for the attenuation of migration. The results suggest that FZS can differentially regulate pathways involved in the migration of OLN93 cells. A deeper insight into the precise role of endogenous FZS would be an essential prerequisite for developing new treatment strategies including supplementation of estradiol and other steroids in preterm infants.
... Finally, plasma concentrations of antioxidant enzymes and glutathione are significantly lower in preterm infants than term infants [6,7]. Thus, the immature brain has a considerably lower antioxidant capacity, which causes oxidative stress for oligodendrocytes' immature stages [8,9] and might lead to a higher risk of intracerebral hemorrhage in preterm infants. ...
... Transient signals can be activated just a few minutes after adding factors and can disappear after minutes or hours [46,47]. E2 and DHEA can induce non-genomic neuroprotective signaling pathways via ERK1/2 and Akt [8,48,49]. ...
... Although it was shown that E2 activates ERK1/2 and Akt signaling to prevent cell death in oligodendrocyte precursor cells [8], we only found downregulation of GSK-3b under DHEA treatment after 6 h in OLN-93 cells. This might be due to differences in cell culture, experimental setting or even the used oligodendrocyte cell type. ...
Article
Full-text available
Preterm birth causes neurological deficits. Previously, we demonstrated that fetal zone steroids reduce hyperoxia-mediated cell death in vitro. In immature oligodendrocytes (OLN-93 cells), dehydroepiandrosterone + 17β-estradiol co-treatment had synergistic beneficial effects while signals were transduced through different receptors. In immature astrocytes (C6 cells), both hormones compete for the same receptor and no synergistic effects were observed. 17β-estradiol and progesterone drastically decrease while fetal zone steroids, mainly dehydroepiandrosterone, remain persistently high within preterm infants until term. Substitution of 17β-estradiol and progesterone does not improve neurological outcomes. We investigated the influence of dehydroepiandrosterone, 17β-estradiol or dehydroepiandrosterone + 17β-estradiol treatment in C6 or OLN-93 cells on steroid receptor availability and activation of intracellular signaling molecules in hyperoxic cell culture. We sought explanations of the observed synergistic effect in preliminary study. In C6 cells, the generated signaling of dehydroepiandrosterone + 17β-estradiol treatment has no synergistic effects. The combined effect on this particular pathway does not potentiate cell survival. In OLN-93 cells, we observed significant differences in the early generated signaling of 17β-estradiol + dehydroepiandrosterone treatment to either 17β-estradiol dehydroepiandrosterone alone but never to both at the same time. The latter finding needs, therefore, further investigation to explain synergistic effects. Nevertheless, we add insight into the receptor and signaling cascade alterations induced by 17β-estradiol, dehydroepiandrosterone or 17β-estradiol + dehydroepiandrosterone treatment of C6 and OLN-93 cells in hyperoxia.
... E2 protects OPCs from hyperoxiainduced apoptosis by downregulating paired immunoglobulin-like receptor B [72] and preventing their migration [95], thereby reducing microglial activation and oxidative stress [19]. These properties result from the activation of E2 receptors and crosstalk with intracellular signaling pathways [138]. However, other studies have found no protective effects of E2 in mouse C8-D1A astrocytes exposed to hyperoxia [139]. ...
Article
Full-text available
The impact of hyperoxia-induced brain injury in preterm infants is being increasingly investigated. However, the parameters and protocols used to study this condition in animal models lack consistency. Research is further hampered by the fact that hyperoxia exerts both direct and indirect effects on oligodendrocytes and neurons, with the precise underlying mechanisms remaining unclear. In this article, we aim to provide a comprehensive overview of the conditions used to induce hyperoxia in animal models of immature brain injury. We discuss what is known regarding the mechanisms underlying hyperoxia-induced immature brain injury, focusing on the effects on oligodendrocytes and neurons, and briefly describe therapies that may counteract the effects of hyperoxia. We also identify further studies required to fully elucidate the effects of hyperoxia on the immature brain as well as discuss the leading therapeutic options.
... Ircariin, which is a flavonoid glycoside, exerts a neuroprotective effect both in P7 mice following the Vannucci's model of HI and in a mouse hippocampal neuronal cell line (HT22) during OGD by activating ERα and reducing apoptosis [25]. When P6 rat pups or the immature oligodendroglia cell line (OLN-93) is exposed to hypoxia estradiol prevents neonatal oxygen-induced white matter injury [26]. It has been suggested that rather than a ligand-based concentration effect, differential ERα nongenomic signaling appears to play the most important role in mediating neuroprotective actions of estradiol in cerebral ischemia [21]. ...
Article
Full-text available
Background Neonatal hypoxia ischemia (HI) related brain injury is one of the major causes of learning disabilities and memory deficits in children. In both human and animal studies, female neonate brains are less susceptible to HI than male brains. Phosphorylation of the nerve growth factor receptor TrkB has been shown to provide sex-specific neuroprotection following in vivo HI in female mice in an estrogen receptor alpha (ERα)-dependent manner. However, the molecular and cellular mechanisms conferring sex-specific neonatal neuroprotection remain incompletely understood. Here, we test whether female neonatal hippocampal neurons express autonomous neuroprotective properties and assess the ability of testosterone (T) to alter this phenotype. Methods We cultured sexed hippocampal neurons from ERα +/+ and ERα −/− mice and subjected them to 4 h oxygen glucose deprivation and 24 h reoxygenation (4-OGD/24-REOX). Sexed hippocampal neurons were treated either with vehicle control (VC) or the TrkB agonist 7,8-dihydroxyflavone (7,8-DHF) following in vitro ischemia. End points at 24 h REOX were TrkB phosphorylation (p-TrkB) and neuronal survival assessed by immunohistochemistry. In addition, i n vitro ischemia-mediated ERα gene expression in hippocampal neurons were investigated following testosterone (T) pre-treatment and TrkB antagonist therapy via q-RTPCR. Multifactorial analysis of variance was conducted to test for significant differences between experimental conditions. Results Under normoxic conditions, administration of 3 µM 7,8-DHF resulted an ERα-dependent increase in p-TrkB immunoexpression that was higher in female, as compared to male neurons. Following 4-OGD/24-REOX, p-TrkB expression increased 20% in both male and female ERα +/+ neurons. However, with 3 µM 7,8-DHF treatment p-TrkB expression increased further in female neurons by 2.81 ± 0.79-fold and was ERα dependent. 4-OGD/24-REOX resulted in a 56% increase in cell death, but only female cells were rescued with 3 µM 7,8-DHF, again in an ERα dependent manner. Following 4-OGD/3-REOX, ERα mRNA increased ~ 3 fold in female neurons. This increase was blocked with either the TrkB antagonist ANA-12 or pre-treatment with T. Pre-treatment with T also blocked the 7,8-DHF- dependent sex-specific neuronal survival in female neurons following 4-OGD/24-REOX. Conclusions OGD/REOX results in sex-dependent TrkB phosphorylation in female neurons that increases further with 7,8-DHF treatment. TrkB phosphorylation by 7,8-DHF increased ERα mRNA expression and promoted cell survival preferentially in female hippocampal neurons. The sex-dependent neuroprotective actions of 7,8-DHF were blocked by either ANA-12 or by T pre-treatment. These results are consistent with a model for a female-specific neuroprotective pathway in hippocampal neurons in response to hypoxia. The pathway is activated by 7,8-DHF, mediated by TrkB phosphorylation, dependent on ERα and blocked by pre-exposure to T.
... 45 E2 also offers protection again oxygen-induced apoptosis of oligodendrocytes and reverses hypoxia-as well as hyperoxia-induced hypomyelination. 46,47 In addition, our previous studies have shown that oestrogen treatment restores interneuron neurogenesis in preterm newborns by cell cycle inhibition and elevation of Ascl1. 48 Hence, E2 functions at multiple levels and in a diverse manner in brain, offering neuroprotection to both neuronal and oligodendrocytic progenitors. ...
Article
Prematurely-born infants cared for in the neonatal units suffer from memory and learning deficits. Prematurity diminishes neurogenesis and synaptogenesis in the hippocampal dentate gyrus (DG). This dysmaturation of neurons is attributed to elevated PSD95, NMDR2A, and IGF1 levels. Since oestrogen treatment plays key roles in the development and plasticity of DG, we hypothesized that 17β-estradiol (E2) treatment would ameliorate neurogenesis and synaptogenesis in the DG, reversing cognitive deficits in premature newborns. Additionally, E2-induced recovery would be mediated by IGF1 signalling. These hypotheses were tested in a rabbit model of prematurity and nonmaternal care, in which premature kits were gavage-fed and reared by laboratory personnel. We compared E2- and vehicle-treated preterm kits for morphological, molecular, and behavioural parameters. We also treated kits with oestrogen degrader, RAD1901, and assessed IGF1 signalling. We found that E2 treatment increased the number of Tbr2+ and DCX+ neuronal progenitors and increased the density of glutamatergic synapses in the DG. E2 treatment restored PSD95 and NMDAR2A levels and cognitive function in preterm kits. Transcriptomic analyses showed that E2 treatment contributed to recovery by influencing interactions between IGF1R and neurodegenerative, as well as glutamatergic genes. ERα expression was reduced on completion of E2 treatment at D7, followed by D30 elevation. E2-induced fluctuation in ERα levels was associated with a reciprocal elevation in IGF1/2 expression at D7 and reduction at D30. ERα degradation by RAD1901 treatment enhanced IGF1 levels, suggesting ERα inhibits IGF1 expression. E2 treatment alleviates the prematurity-induced maldevelopment of DG and cognitive dysfunctions by regulating ERα and IGF1 levels.
... After GAS5 has been downregulated in RA, the expression of HDAC4 in the cell is inhibited, thereby increasing the phosphorylation level of protein kinase B (AKT) in the AKT/mammalian target of rapamycin (mTOR) signaling pathway. In addition, there is already evidence that activated Akt Bad, a member of the proapoptotic B-cell lymphoma-2 (Bcl-2) family, phosphorylates and induces its inactivation (56). Therefore, downregulation of LncRNA GAS5 can promote cell apoptosis. ...
Article
Full-text available
Rheumatoid arthritis (RA) is a systemic chronic autoinflammatory disease, and the synovial hyperplasia, pannus formation, articular cartilage damage and bone matrix destruction caused by immune system abnormalities are the main features of RA. The use of Disease Modifying Anti-Rheumatic Drugs (DMARDs) has achieved great advances in the therapy of RA. Yet there are still patients facing the problem of poor response to drug therapy or drug intolerance. Current therapy methods can only moderate RA progress, but cannot stop or reverse the damage it has caused. Recent studies have reported that there are a variety of long non-coding RNAs (LncRNAs) that have been implicated in mediating many aspects of RA. Understanding the mechanism of LncRNAs in RA is therefore critical for the development of new therapy strategies and prevention strategies. In this review, we systematically elucidate the biological roles and mechanisms of action of LncRNAs and their mechanisms of action in RA. Additionally, we also highlight the potential value of LncRNAs in the clinical diagnosis and therapy of RA.
... Indeed, for all other tracts of interest, a three-way interaction did often trend in the same direction, and towards significance (see supplemental materials). Regarding potential mechanisms for the sex interaction, there is preliminary evidence from cellular animal models that estrogen may play a critical role in myelinogenesis [44], and attenuate damage secondary to pathology or neural injury [45]. Though research examining protective mechanisms of estrogen in humans is limited, one study has found that estrogen deficiency in girls with Turner Syndrome is linked to significant aberrations in white matter, particularly reduced FA in the SLF in comparison to TD youth [46]. ...
Article
Full-text available
Pediatric post-traumatic stress disorder (pPTSD) is a prevalent and pervasive form of mental illness comprising a disparate constellation of psychiatric symptoms. Emerging evidence suggests that pPTSD may be characterized by alterations in functional networks traversing the brain. Yet, little is known about pathological changes in the structural tracts underlying functional connectivity. In adults, PTSD is linked to widespread change in white matter integrity throughout the brain, yet similar studies with youth populations have yet to be conducted. Current understanding of the nature and treatment of pPTSD may be enhanced by examining alterations in white matter, while further untangling effects of age and sex. Here, we assess the microstructure of 12 major white matter tracts in a sample of well-phenotyped youth with PTSD. Measures of fractional anisotropy were derived from diffusion tensor images acquired from 82 unmediated youth (ages 8–18), of whom 39 met criteria for pPTSD. Diagnosis of pPTSD was linked to remarkable age- and sex-linked differences in the microstructure of major white matter tracts including the uncinate fasciculus, cingulum bundle, and inferior longitudinal fasciculus. In each case, youth with PTSD show an absence of increased white matter integrity with age, suggesting an altered pattern of neurodevelopment that may contribute to persistence or worsening of illness. Broadly, our results suggest abnormal white matter development in pediatric PTSD, a finding which may contribute to illness persistence, comorbidity with other disorders, and poorer prognosis across time. Critically, these findings further speak to the nature of pPTSD as a ‘whole-brain’ disorder.
... The presence of high amounts of estrogen is considered to be critical for normal brain development [8,9]. Several experimental studies have highlighted that the female sex hormones represent potential neuroprotective agents against acute and chronic injuries in the brain [10][11][12]. But at the time of birth, after the umbilical cord is clamped, E2 level decreases by a factor of 100 within 24 h [13]. ...
Article
Full-text available
Oxygen causes white matter damage in preterm infants and male sex is a major risk factor for poor neurological outcome, which speculates the role of steroid hormones in sex-based differences. Preterm birth is accompanied by a drop in 17β-estradiol (E2) and progesterone along with increased levels of fetal zone steroids (FZS). We performed a sex-based analysis on the FZS concentration differences in urine samples collected from preterm and term infants. We show that, in preterm urine samples, the total concentration of FZS, and in particular the 16α-OH-DHEA concentration, is significantly higher in ill female infants as compared to males. Since we previously identified Nup133 as a novel target protein affected by hyperoxia, here we studied the effect of FZS, allopregnanolone (Allo) and E2 on differentiation and Nup133 signaling using mouse-derived primary oligodendrocyte progenitor cells (OPCs). We show that the steroids could reverse the effect of hyperoxia-mediated downregulation of Nup133 in cultured male OPCs. The addition of FZS and E2 protected cells from oxidative stress. However, E2, in presence of 16α-OH-DHEA, showed a negative effect on male cells. These results assert the importance of sex-based differences and their potential implications in preterm stress response.
... Indeed, for all other tracts of interest, a three-way interaction did often trend in the same direction, and towards significance (see supplemental materials). Regarding potential mechanisms for the sex interaction, there is preliminary evidence from cellular animal models that estrogen may play a critical role in myelinogenesis [44], and attenuate damage secondary to pathology or neural injury [45]. Though research examining protective mechanisms of estrogen in humans is limited, one study has found that estrogen deficiency in girls with Turner Syndrome is linked to significant aberrations in white matter, particularly reduced FA in the SLF in comparison to TD youth [46]. ...
Article
Full-text available
Myelin plays a pivotal role in the efficient transmission of nerve impulses. Disruptions in myelin integrity are associated with numerous neurological disorders, including multiple sclerosis. In the central nervous system (CNS), myelin is formed by oligodendrocytes. Remyelination refers to the re-formation of the damaged myelin sheath by newly formed oligodendrocytes. Steroids have gained attention for their potential modulatory effects on myelin in both health and disease. Steroids are traditionally associated with endocrine functions, but their local synthesis within the nervous system has generated significant interest. The term "neuroactive steroids" refers to steroids that can act on cells of the nervous system. In the healthy state, neuroactive steroids promote myelin formation, maintenance, and repair by enhancing oligodendrocyte differentiation and maturation. In pathological conditions, such as demyelination injury, multiple neuroactive steroids have shown promise in promoting remyelination. Understanding the effects of neuroactive steroids on myelin could lead to novel therapeutic approaches for demyelinating diseases and neurodegenerative disorders. This review highlights the potential therapeutic significance of neuroactive steroids in myelin-related health and diseases. We review the synthesis of steroids by neurons and glial cells and discuss the roles of neuroactive steroids on myelin structure and function in health and disease. We emphasize the potential pro-myelinating effects of the varying levels of neuroactive steroids during different female physiological states such as the menstrual cycle, pregnancy, lactation, and post-menopause.
Article
Full-text available
The importance of postmenopausal estrogen replacement therapy in affording protection against the selective and delayed neuronal death associated with cardiac arrest or cardiac surgery in women remains controversial. Here we report that exogenous estrogen at levels that are physiological for hormone replacement in postmenopausal women affords protection against global ischemia-induced neuronal death and prevents activation of apoptotic signaling cascades in the hippocampal CA1 of male gerbils. Global ischemia induced a marked increase in activated caspase-3 in CA1, evident at 6 hr after ischemia. Global ischemia induced a marked upregulation of the proapoptotic neurotrophin receptor p75 NTR in CA1, evident at 48 hr. p75 NTR expression was induced primarily in terminal deoxynucleotidyl transferase-mediated UTP nick-end labeling-positive cells, indicating expression in neurons undergoing apoptosis. Global ischemia also induced a marked downregulation of mRNA encoding the AMPA receptor GluR2 subunit in CA1. Caspase-3, p75 NTR , and GluR2 were not significantly changed in CA3 and dentate gyrus, indicating that the ischemia-induced changes in gene expression were cell specific. Exogenous estrogen attenuated the ischemia-induced increases in activated caspase-3 and blocked the increase in p75 NTR in post-ischemic CA1 neurons but did not prevent ischemia-induced downregulation of GluR2. These findings demonstrate that long-term estrogen at physiological levels ameliorates ischemia-induced hippocampal injury and indicate that estrogen intervenes at the level of apoptotic signaling cascades to prevent onset of death in neurons otherwise “destined to die.”
Article
Full-text available
Hypoxic-ischemic injury to the periventricular cerebral white matter [periventricular leukomalacia (PVL)] results in cerebral palsy and is the leading cause of brain injury in premature infants. The principal feature of PVL is a chronic disturbance of myelination and suggests that oligodendrocyte (OL) lineage progression is disrupted by ischemic injury. We determined the OL lineage stages at risk for injury during the developmental window of vulnerability for PVL (23–32 weeks, postconceptional age). In 26 normal control autopsy human brains, OL lineage progression was defined in parietal white matter, a region of predilection for PVL. Three successive OL stages, the late OL progenitor, the immature OL, and the mature OL, were characterized between 18 and 41 weeks with anti-NG2 proteoglycan, O4, O1, and anti-myelin basic protein (anti-MBP) antibodies. NG2+O4+ late OL progenitors were the predominant stage throughout the latter half of gestation. Between 18 and 27 weeks, O4+O1+ immature OLs were a minor population (9.9 ± 2.1% of total OLs; n = 9). Between 28 and 41 weeks, an increase in immature OLs to 30.9 ± 2.1% of total OLs ( n = 9) was accompanied by a progressive increase in MBP+ myelin sheaths that were restricted to the periventricular white matter. The developmental window of high risk for PVL thus precedes the onset of myelination and identifies the late OL progenitor as the major potential target. Moreover, the decline in incidence of PVL at ∼32 weeks coincides with the onset of myelination in the periventricular white matter and suggests that the risk for PVL is related to the presence of late OL progenitors in the periventricular white matter.
Article
Full-text available
Until recently, steroid hormones were believed to act only on cells containing intracellular receptors. However, recent evidence suggests that steroids have specific and rapid effects at the cellular membrane. Using whole-cell patch-clamp techniques, 17 beta-estradiol was found to reduce Ba2+ entry reversibly via Ca2+ channels in acutely dissociated and cultured neostriatal neurons. The effects were sex-specific, i.e., the reduction of Ba2+ currents was greater in neurons taken from female rats. 17 beta-Estradiol primarily targeted L-type currents, and their inhibition was detected reliably within seconds of administration. The maximum reduction by 17 beta-estradiol occurred at picomolar concentrations. 17 beta-Estradiol conjugated to bovine serum albumin also reduced Ba2+ currents, suggesting that the effect occurs at the membrane surface. Dialysis with GTP gamma S prevented reversal of the modulation, suggesting that 17 beta-estradiol acts via G-protein activation. 17 alpha-Estradiol also reduced Ba2+ currents but was significantly less effective than 17 beta-estradiol. Estriol and 4-hydroxyestradiol were found to reduce Ba2+ currents with similar efficacy to 17 beta-estradiol, whereas estrone and 2-methoxyestriol were less effective. Tamoxifen also reduced Ba2+ currents but did not occlude the effect of 17 beta-estradiol. These results suggest that at physiological concentrations, 17 beta-estradiol can have immediate actions on neostriatal neurons via nongenomic signaling pathways.
Article
Full-text available
A new method of total RNA isolation by a single extraction with an acid guanidinium thiocyanate-phenol-chloroform mixture is described. The method provides a pure preparation of undegraded RNA in high yield and can be completed within 4 h. It is particularly useful for processing large numbers of samples and for isolation of RNA from minute quantities of cells or tissue samples.
Article
Full-text available
Infants born very preterm have an increased risk of brain injury. Given the great increase in the number of such infants that are surviving, it is important to establish whether any resultant brain abnormalities persist into adolescence and adult life. We therefore examined in vivo whole brain, grey matter, white matter and hip-pocampal volumes, ventricular size and grey/white matter ratios in a series of adolescents who had been born very preterm, and an age-matched full-term control group. Structural MRI was carried out on a cohort of 72 adolescents (mean age 15 years) who were born before 33 weeks, and 48 age-matched full-term controls. Brain measurements were made blind to group af®li-ation using stereological principles. After controlling for gender and height, the very preterm subjects showed a 6.0% decrease in whole brain volume, and an 11.8% decrease in cortical grey matter volume, as well as a 15.6% decrease in right and a 12.1% decrease in left hippocampal volumes; they also had a 42.0% increase in the size of the lateral ventricles. Therefore, individuals who were born very preterm continue to show noticeable decrements in brain volumes and striking increases in lateral ventricular volume into adolescence. The functional signi®cance of these abnormalities merits further investigation.
Article
Although very-low-birth-weight (VLBW) infants, those weighing less than 1500 g, gained significantly better outcomes when neonatal intensive care became available in the 1960s, studies of later mental development have given mixed results. This longitudinal study enrolled 242 VLBW infants born in the years 1977-1979 who survived and 233 control infants of normal birth weight from the same population. The mean gestational age of the VLBW group at birth was 29.7 weeks. Intelligence testing was performed at age 20 with the Vocabulary and Block Design subtests of the Short Form of the Wechsler Adult Intelligence Scale-Revised. These tests measure verbal comprehension and perceptual-organizational skills, respectively. Academic skills were evaluated using the Letter-Word Identification and Applied Problems subtests of the Woodcock-Johnson Psycho-Educational Battery-Revised. The VLBW and normal-weight groups were sociodemographically similar, but the former group had significantly more chronic conditions than did control group. The major differences were in neurosensory impairment and subnormal height. Fewer subjects in the VLBW group had graduated from high school or obtained a general equivalency diploma by age 20. Although 40% of the VLBW group had repeated a grade, the figure for control subjects was 27%. Fewer VLBW subjects were enrolled in post-secondary studies. These differences remained significant when analysis was limited to those of normal IQ who had no neurosensory impairment. Mean IQ scores were significantly lower in the VLBW group (87 vs. 92, P <.001), and these subjects more often had an IQ less than 70. Control subjects had used alcohol and marijuana more than VLBW subjects, and they more often had had contact with the police. Fewer VLBW subjects had had intercourse, been pregnant, or delivered a live-born infant. Neurodevelopmental expressions of very low birth weight and underachievement at school persist at least into young adulthood. In this study, however, these problems were not associated with increased risk-taking behavior or criminal activity.
Article
The plasma concentrations of progesterone, 17-hydroxyprogesterone, and unconjugated estrone, estradiol, and estriol, were measured by radioligand assays in 0.21 ml. plasma aliquots obtained during the second half of normal human pregnancy. The mean plasma concentrations of unconjugated estrone, estradiol, and estriol as well as progesterone gradually increased toward term whereas the mean plasma 17-hydroxyprogesterone concentrations increased significantly only after the thirty-third week of gestation. Mean plasma progesterone concentrations were at least ninefold higher than any of the unconjugated estrogens measured in this study. Estradiol was quantitatively the most important unconjugated estrogen throughout the second half of pregnancy. The ratios of plasma progesterone: estradiol (unconjugated), progesterone: estriol (unconjugated), and estradiol (unconjugated): estriol (unconjugated) remained constant throughout the second half of gestation. There appears to be no preferential increase in the plasma concentrations of any one of these hormones as measured.
Article
Background and Purpose—Recent evidence suggests that endogenous estrogens or hormone replacement therapy can ameliorate brain damage from experimental stroke. Protective mechanisms involve enhanced cerebral vasodilation during ischemic stress as well as direct preservation of neuronal viability. We hypothesized that if the intracellular estrogen receptor subtype-α (ERα) is important to estrogen’s signaling in the ischemic brain, then ERα-deficient (knockout) (ERαKO) female mice would sustain exaggerated cerebral infarction damage after middle cerebral artery occlusion. Methods—The histopathology of cresyl violet–stained tissues was evaluated after reversible middle cerebral artery occlusion (2 hours, followed by 22 hours of reperfusion) in ERαKO transgenic and wild-type (WT) mice (C57BL/6J background strain). End-ischemic cerebral blood flow mapping was obtained from additional female murine cohorts by using [¹⁴C]iodoantipyrine autoradiography. Results—Total hemispheric tissue damage was not altered by ERα deficiency in female mice: 51.9±10.6 mm³ in ERαKO versus 60.5±5.0 mm³ in WT. Striatal infarction was equivalent, 12.2±1.7 mm³ in ERαKO and 13.4±1.0 mm³ in WT mice, but cortical infarction was paradoxically smaller relative to that of the WT (20.7±4.5 mm³ in ERαKO versus 30.6±4.1 mm³ in WT). Intraocclusion blood flow to the parietal cortex was higher in ERαKO than in WT mice, likely accounting for the reduced infarction in this anatomic area. There were no differences in stroke outcomes by region or genotype in male animals. Conclusions—Loss of ERα does not enhance tissue damage in the female animal, suggesting that estrogen inhibits brain injury by mechanisms that do not depend on activation of this receptor subtype.