ArticlePDF Available

Lemaire V, Koehl M, Le Moal M, Abrous DN. Prenatal stress produces learning deficits associated with an inhibition of neurogenesis in the hippocampus. Proc Natl Acad Sci USA 97: 11032-11037

Authors:

Abstract and Figures

Early experiences such as prenatal stress significantly influence the development of the brain and the organization of behavior. In particular, prenatal stress impairs memory processes but the mechanism for this effect is not known. Hippocampal granule neurons are generated throughout life and are involved in hippocampal-dependent learning. Here, we report that prenatal stress in rats induced lifespan reduction of neurogenesis in the dentate gyrus and produced impairment in hippocampal-related spatial tasks. Prenatal stress blocked the increase of learning-induced neurogenesis. These data strengthen pathophysiological hypotheses that propose an early neurodevelopmental origin for psychopathological vulnerabilities in aging.
Content may be subject to copyright.
Prenatal stress produces learning deficits associated
with an inhibition of neurogenesis in
the hippocampus
V. Lemaire*, M. Koehl*, M. Le Moal, and D. N. Abrous
Laboratoire de Psychobiologie des Comportement Adaptatifs, Institut National de la Sante´ et de la Recherche Me´ dicale Unit 259, Universite´ de Bordeaux II,
Domaine de Carreire, Rue Camille Saint Sae¨ ns, 33077 Bordeaux Cedex, France
Edited by Mark R. Rosenzweig, University of California, Berkeley, CA, and approved July 7, 2000 (received for review March 21, 2000)
Early experiences such as prenatal stress significantly influence the
development of the brain and the organization of behavior. In
particular, prenatal stress impairs memory processes but the mech-
anism for this effect is not known. Hippocampal granule neurons
are generated throughout life and are involved in hippocampal-
dependent learning. Here, we report that prenatal stress in rats
induced lifespan reduction of neurogenesis in the dentate gyrus
and produced impairment in hippocampal-related spatial tasks.
Prenatal stress blocked the increase of learning-induced neurogen-
esis. These data strengthen pathophysiological hypotheses that
propose an early neurodevelopmental origin for psychopatholog-
ical vulnerabilities in aging.
I
t is well documented from animal studies that during the perinatal
period, the development of an organism is subjected to complex
environmental influences. Deleterious life events during pregnancy
induce neurobiological and behavioral defects in offspring, some of
them involving the hippocampal formation (1–5). Indeed, prenatal
stress results in an enhanced production of stress hormones by the
mother during critical periods of fetal brain development and
provokes a definitively longer corticosterone response to stress in
the offspring associated with a reduction in the number of hip-
pocampal corticosteroid receptors (1, 3, 5). Behaviorally, the prog-
eny, from adulthood to senescence, exhibit memory deficits in a
hippocampal-dependent task (2, 4, 5).
Recently, it has been hypothesized that hippocampal-mediated
learning (6) may be related to the generation of new neurons in the
adult dentate gyrus (7, 8, 9). These newborn cells migrate in the
granule cell layer, and differentiate in granule neurons whose
projections, the mossy fibers, extend to the CA3 hippocampal
region (10, 11). Furthermore, the size of the mossy fibers’ projec-
tions correlates with variations in performances in spatial memory
tests (12, 13). Finally, glucocorticoid levels regulate de novo cell
proliferation in the dentate gyrus. Indeed, adrenalectomy per-
formed in young or aged rats increases neurogenesis, an effect that
is prevented by glucocorticoid treatment (14, 15, 16). These results
raised the critical question as to whether prenatal stress can impair
neurogenesis and, if so, whether it is related to learning ability.
To test this hypothesis, we first examined cell proliferation in
the progeny of stressed mothers with 5-bromo-2-deoxyuridine
(BrdUrd), a thymidine analogue incorporated into genetic material
during synthetic DNA phase (S phase) of mitotic division. Cell-
specific markers were used to phenotype the newly born neurons
after longer survival times. We next examined whether the struc-
tural hippocampal defects resulting from prenatal stress had func-
tional consequences on learning abilities. Finally, we examined
whether the reduction in cell proliferation in the dentate gyrus had
an impact on spatial memory, a hypothesis supported by others (17).
Methods
Housing Conditions. Adult virgin Sprague–Dawley female rats (Iffa
Credo) weighing 240 g were housed, 10 days after arrival, in the
presence of a sexually experienced male Sprague–Dawley rat
weighing 400 g. Pregnant females then were randomly assigned to
prenatal stress (PS) or control (C) groups and individually housed
in plastic breeding cages. Animals were allowed ad libitum access
to food and water and were maintained on a constant lightdark
cycle with constant temperature and humidity.
Prenatal Stress. Stress was performed each day of the last week of
pregnancy from day 15 until delivery. Pregnant females were
individually restrained for 45 min three times a day during the
light phase in plastic transparent cylinders (7-cm diameter,
19-cm long) exposed to bright light. Control pregnant females
were left undisturbed in their home cages. The offspring were
raised by their biological mothers until weaning (21 days after
birth). Only litters of 8–13 pups with similar numbers of males
and females were kept for the study, all other litters having been
eliminated to rule out additional stressors such as removal of the
pups. For each set of experiments, a maximum of two male pups
was taken from each litter to remove any ‘‘litter effects’’ (18).
After weaning, male rats from each experimental group (PS, n
39; C, n 34) were housed in groups of three. One month before
each behavioral study, animals were individually housed.
Weight of Adrenal Glands. To have an index of chronic hypo-
thalamic–pituitary–adrenal (HPA) axis activity (19, 20), adrenal
glands were collected on the day of the animals were killed. They
were immediately weighed and results are expressed as a ratio of
body weight.
Place Navigation Task. Four-month-old rats (C, n 10; PS, n 10)
were used in this experiment. The apparatus consisted of a circular
swimming pool built of gray plastic (180-cm diameter 60-cm
high), which was filled with water at room temperature and made
opaque by the addition of milk. In this task, animals are required
to locate a platform submerged (1.5 cm) in the pool by using only
the spatial cues available within the testing room. During the
habituation phase (no platform), rats (C, n 5; PS, n 5) were
given one trial per day (of 60 sec) over 3 days. During the testing
phase, the platform was hidden in a fixed location in one of four
quadrants halfway between the sidewalls and the center of the pool.
The animals were given four trials per day (90 sec) over 5 days. As
a control of training, rats of both groups (C, n 5; PS, n 5) were
handled in parallel with the trained rats but were not submitted to
the water maze task (manipulated groups).
This paper was submitted directly (Track II) to the PNAS office.
Abbreviations: PS group, prenatal stress group; C group, control group; HPA, hypothalam-
ic–pituitary–adrenal; IR, immunoreactivity; GFAP, glial fibrillary acidic protein; NeuN, neu-
ronal nuclei; n.s., not significant.
*V.L. and M.K. contributed equally to this work.
To whom reprint requests should be addressed. E-mail: Nora.Abrous@bordeaux.inserm.fr.
The publication costs of this article were defrayed in part by page charge payment. This
article must therefore be hereby marked advertisement in accordance with 18 U.S.C.
§1734 solely to indicate this fact.
11032–11037
PNAS
September 26, 2000
vol. 97
no. 20
BrdUrd Injections. Rats were injected with BrdUrd dissolved in
phosphate buffer (21). Animals of different ages received four
injections: once 3 days before death, once 2 days before death,
and twice the day before death (15, 16). To determine the
phenotype of the newly born cells, 3-month-old control and
prenatally stressed rats were killed 2 weeks after the last BrdUrd
injection. For the behavioral study, 4-month-old animals trained
in the water maze were injected once a day, 2 min before the first
trial during the third, fourth, and fifth days of training. The
manipulated groups were injected in parallel. Animals were
killed the day after the last injection.
Histological Procedure. Rats were deeply anesthetized with chloral
hydrate (400 mgkg i.p.) and were perfused with 150 ml of
phosphate buffer saline (pH, 7.3) containing heparin (5 10
4
unitsml), followed by 300 ml of 4% paraformaldehyde in 0.1 M
of phosphate buffer (pH, 7.3). After a 24-h postfixation period
of the brains in paraformaldehyde, 50-
m frontal sections were
cut on a vibratome and collected in PBS (0.1 M; pH, 7.4).
Free-floating sections were processed in a standard immunohis-
tochemical procedure (15, 16, 22). For BrdUrd labeling, sections
were treated with 2 M HCl (30 min at 37°C) and then rinsed in
borate buffer for 5 min (0.1 M; pH, 8.4). They were extensively
washed with PBS, preincubated 45 min with PBS containing
0.3% Triton X-100 and 3% of horse normal serum (blocking
solution), and incubated under agitation for 72 h at 4°C in mouse
monoclonal anti-BrdUrd antibody (1200; Dako) diluted in PBS
containing 0.3 Triton X-100 and 1% of horse normal serum. The
sections were then incubated under agitation for2hwitha
biotin-labeled horse anti-mouse IgG antibody (1200; Vector
Valbiotec, Paris). Sections from all animals were processed in
parallel and immunoreactivities (IRs) were visualized by the
biotin–streptavidin technique (ABC kit; Dako) with 3,3-
diaminobenzidine as chromogen (10-min incubation).
The phenotype of newly born cells was examined by double-
labeled immunohistofluorescence according to a previously de-
scribed method (22). Briefly, sections were incubated with a rabbit
polyclonal anti-glial fibrillary acidic protein (GFAP) antibody (1
10,000; Dako) or with a mouse monoclonal anti-neuronal nuclei
(NeuN) antibody (11,000; Chemicon). Bound anti-GFAP or
-NeuN antibodies were visualized respectively with an Alexa 488
goat anti-rabbit IgG antibody and a Alexa goat anti-mouse IgG
antibody (11,000; Interchem, Montluc¸on, France). Then, sections
were incubated with a rat monoclonal anti-BrdUrd antibody (1
1,000; Accurate Scientific, Westbury, NY), and bound anti-BrdUrd
molecules were revealed by using a Cy3-labeled anti-rat IgG
antibody (1400; Jackson ImmunoResearch).
Quantitative Evaluation of Staining. For each animal, starting ran-
domly, BrdUrd-IR cells were counted in the left dorsal hippocam-
pus, in one in five sections, 250
m apart. All BrdUrd-IR cells were
counted with a 100 microscope objective in the subgranular layer
and in the granular layer of the dentate gyrus. The sections then
were counterstained and the surface of the granule cell layer was
measured with a Samba 2640 system (Alcatel system; TITN An-
sware, Grenoble, France). For each section, the numerical density
of BrdUrd-IR was calculated by dividing the number of BrdUrd-IR
cells by granule cell layer sectional volume. For each animal, the
mean numerical density of BrdUrd-IR was calculated. The total
number of BrdUrd-IR cells per dentate gyrus then was calculated
by multiplying the numerical density of BrdUrd-IR cells by the
reference volume. The volume of the reference space was calcu-
lated according to the Cavalieri estimation (23) applying the
following formula: V
ref
a t s, where a is the mean area of
the granule cell layer of the dentate gyrus, t is the thickness of the
vibratome section (50
m), and s is the total number of sections
through the defined regions of the dorsal hippocampus. The
number of pyknotic cells was determined on hematoxylin-
counterstained sections by using a similar procedure.
The absolute number of granule cells, as assessed morpho-
logically by hematoxylin staining, was determined by using a
Samba 2640 system. For each section, granule cells were counted
in 20 to 25 frames (20
m 20
m) at evenly spaced x–y intervals
of 110
mby110
m. We disregarded cells that were in sharp
focus in the uppermost focal plane (optical disector principle).
The numerical density (number of cellsmm
3
) was calculated
according to the following equation:
N
v
Q
h afra
P
, [1]
where (Q
) is the sum of cells, (P) the number of frame, (h) the
disector height, and a(fra) the area of the counting frame. By using
these sampling parameters, the number of optical disectors exam-
ined was 53.67 1.10 [CE(Fi), 13.67 0.22%], the number of cells
counted per dentate gyrus was 221.32 7.15 [CE(Qi), 13.99
0.25%], and the CE(N
V
) was 13.69 0.22% (CE is the coefficient
of error). The total number of cells then was calculated by multi-
plying the numerical density by the volume of reference (ref. 23 and
see above).
General Procedures. First experiment: Prenatal stress and cell pro-
liferation. Juvenile, adult, middle-aged, and old rats (C, n 20;
PS, n 25) were killed the day after the last BrdUrd injection.
These rats were considered ‘‘naive’’ as they were not manipu-
lated except for injections and were not behaviorally tested. The
Fig. 1. Effect of prenatal stress on cell
proliferation in 28-day-, 3-month-, 10-
month-, and 22-month-old male rats. There
was a significant decline of cell proliferation
with increasing age in both control and pre-
natally stressed rats. Furthermore, for the
four ages investigated, prenatal stress consis-
tently decreased cell proliferation as com-
pared with control.
*
, P 0.05 in comparison
to age-matched control group;
**
, P 0.01.
Notice the different ordinal scale between
data for 28-day-old rats and the other ages
studied.
Lemaire et al. PNAS
September 26, 2000
vol. 97
no. 20
11033
NEUROBIOLOGY
total number of BrdUrd-IR cells per dentate gyrus was counted
on diaminobenzidine-immunostained sections. The absolute
number of granule neurons was determined on the same sections
after counterstaining. The total number of pyknotic cells was
determined on alternate counterstained sections.
Second experiment: Phenotype of the newly born cells. Adult
naive rats (C, n 4; PS, n 4) were injected four times with
BrdUrd and were killed 2 weeks after the last BrdUrd injection.
The total number of BrdUrd-IR cells per dentate gyrus was
counted on diaminobenzidine-immunostained sections. Double
immunofluorescence was performed and the identity of newly
born cells was examined with cell-specific markers.
Third experiment: Training in the water maze and cell prolifer-
ation.
Four-month-old rats were either trained in the water maze
(trained groups: C, n 5; PS, n 5) or handled in parallel with
the trained rats but not submitted to the water maze task
(manipulated groups: C, n 5; PS, n 5). Rats were injected
four times with BrdUrd (as described above) and were killed the
day after the last injection. The total number of BrdUrd-IR cells
per dentate gyrus was counted on diaminobenzidine-
immunostained sections. The absolute number of granule neu-
rons was determined on the same sections after counterstaining.
Results
Effect of Prenatal Stress on Cell Proliferation. We first examined, in
the progeny of stressed mothers, neurogenesis in the dentate gyrus,
a process known to be regulated by corticosterone levels (14, 15,
16). We also examined whether cell proliferation was affected at
specific times during the lifespan, including in senescent rats. Cell
proliferation was evaluated in rats injected with BrdUrd; cells that
incorporated BrdUrd (BrdUrd-IR cells) were revealed by immu-
nohistochemistry. BrdUrd-IR cell number was determined in ju-
venile (28-day-old), adult (3-month-old), middle-aged (10-month-
old), and old (22-month-old) naive rats (Figs. 1 and 2). There was
a significant decline of proliferation as age increased in both control
[H(3, 19) 16.75, P 0.001; 28 days, 3 months, 10 months, 22
months at P 0.05 with a nonparametric Kruskal–Wallis ANOVA]
and prenatally stressed rats [H(3, 25) 22.09, P 0.001; 28 days,
3 months, 10 months, 22 months at P 0.01]. Furthermore,
for the four ages investigated, prenatal stress consistently decreased
cell proliferation {38.4% at 28 days [H(1, 11) 4.03, P 0.05];
59.3% at 3 months [H(1, 11) 7.51, P 0.01]; 42.3% at 10
months [H(1, 12) 6.56, P 0.01]; and 55.2% at 22 months of
age [H(1, 10) 4.69; P 0.05]}. These differences were not related
Fig. 2. Illustration of BrdUrd-labeled cells in the dentate gyrus. Photomicrographs of BrdUrd immunoreactivity in frontal section of the dentate gyrus in a
control (a) and a prenatally stressed (b) rat . (Scale 1.5 cm for 500
m.) (c) Confocal illustration of BrdUrd-labeled cells in the dentate gyrus showing that newborn
cells were mainly neurons. Indeed, BrdUrd-IR cells (red nuclear stain, Cy3) colocalized with the neuronal marker NeuN (green stain) as indicated by the yellow
nuclear staining. (d) Newly born cells are rarely astrocytes because BrdUrd-IR cells (red nuclear stain) is not stained for the astroglial marker GFAP (green stain).
11034
www.pnas.org Lemaire et al.
to differences in the volume of the granule cell layer [28 days: H(1,
11) 0.33, not significant (n.s.); 3 months: H(1, 11) 2.13, n.s.; 10
months: H(1, 12) 0.10, n.s.; 22 months: H(1, 10) 0.64, n.s.].
Identity of Newly Born Cells. To examine the phenotype of Br-
dUrd-IR cells born in the dentate gyrus of control (Fig. 2a)or
prenatally stressed (Fig. 2b) rats, animals were allowed to survive
for 2 weeks after the last BrdUrd injection. Newborn cells, pheno-
typed by analysis of double-labeled immunofluorescence sections,
were mainly neurons and rarely astrocytes (Fig. 2 c and d). Indeed,
72 2% and 70 2% of BrdUrd-IR cells were immunoreactive for
NeuN in control and prenatally stressed rats, respectively. These
ratios were not significantly different among groups [H(1, 8) 0.08;
n.s.]. On the other hand, 20 1.6% and 20 1.6% of BrdUrd-IR
cells were immunoreactive for GFAP in control and prenatally
stressed rats, respectively. These ratios were not significantly dif-
ferent among groups [H(1, 8) 2.08, n.s.]. The double labeling was
verified with confocal microscopy. Exploration in the Z plane
revealed that bound anti-GFAP antibodies were present through-
out the section thickness, but bound anti-NeuN antibodies pene-
trate only a portion of the slice. Thus, the quantification of
BrdUrd-NeuN double-labeled cells was underestimated.
Effect of Prenatal Stress on the Total Number of Granule Neurons.
Because prenatal stress reduces cell proliferation in the dentate
gyrus early in life and in aged rats, it was of interest to evaluate the
consequence of this reduction on the total granule cell number.
Although the total number of granule neurons was not different
among different ages in control rats [H(3, 20) 6.61, n.s.] (Fig. 3),
there was a progressive decline of total granule cell number with
increasing age after prenatal stress [H(3, 25) 14.24, P 0.01; 28
days, 3 months, 10 months, 22 months at P 0.01]. Prenatal
stress decreased the total number of granule neurons beginning at
3 months of age [H(1, 11) 4.80, P 0.05] and continued until 10
[H(1, 12) 4.33, P 0.05] and 22 [H(1, 10) 4.69, P 0.05]
months of age.
The proportion of newborn cells in the dentate gyrus was
computed in percentage of total granule cells and represented
5.8% 0.7 in 28-day-old control rats (the largest percentage) to
0.08% 0.02 in 22-month-old prenatally stressed rats (the
lowest percentage). Thus, the low proportion of newborn cells
could explain why the deficit in cell proliferation is detectable
before the decrease in the total number of granule cells in
prenatally stressed rats.
Effect of Prenatal Stress on the Total Number of Pyknotic Cells. To
determine whether the observed alteration of granule neurons
number was related specifically to differences in cell proliferation,
we evaluated the consequence of prenatal stress on pyknotic cell
number (Fig. 4). The degenerating profiles were characterized by a
condensed chromatin and a light or absent cytoplasm. Prenatal
stress did not influence the total number of pyknotic cells at all ages
tested [3 months, H(1, 11) 0.80, n.s.; 10 months, H(1, 11) 0.38,
n.s.; 22 months, H(1, 10) 0.11, n.s.]. In contrast, there was an
age-related increase in degenerating profile in both control [H(2,
12) 9.9, P 0.01; 3 months, 10 months, 22 months at P
0.01] and prenatally stressed [H(2, 19) 22.09, P 0.01; 3 months,
10 months, 22 months at P 0.01] rats.
HPA Axis Activity. The weight of adrenal glands (expressed as a
ratio of the body weight) previously has been demonstrated to be
a reliable index of the chronic activity of the HPA axis because
a hypertrophy of the adrenal glands is observed in conditions of
chronic HPA axis hyperactivity (19, 20). As shown on Table 1,
this ratio was higher in prenatally stressed rats when compared
with control rats [H(1, 11) 4.03, P 0.05; H(1, 11) 7.50, P
0.01; H(1, 12) 3.69, P 0.05)] indicating a chronic up-
regulation of the HPA axis.
Effect of Prenatal Stress on Spatial Learning. We next examined
whether such structural hippocampal defects resulting from
prenatal stress had functional correlates on spatial memory and
whether cell proliferation was influenced by training. We tested
4-month-old rats for spatial memory in a water maze.
Rats were injected with BrdUrd before the testing session on
days 3, 4, and 5 of learning. At 24 h after the last BrdUrd
injection, all animals were perfused and cell proliferation was
determined. There were significant differences in the rate of
acquisition between control and prenatally stressed rats (Fig. 5).
The latency to find the platform by the control group was
significantly less than that observed for the prenatally stressed
group [H(1, 10) 6.82, P 0.001]. The distance covered by
control animals was shorter than that covered by prenatally
stressed rats (data not shown, P 0.05).
As shown in Fig. 6, acquisition of this spatial task increased
hippocampal neurogenesis in control rats [H(1, 10) 4.81, P
0.05] but not in prenatally stressed rats [H(1, 10) 0.88, n.s.]. As
previously shown, prenatal stress decreased the total number of
Fig. 3. Effect of prenatal stress on total number of granule cells in 28-day-,
3-month-, 10-month-, and 22-month-old male rats. Although the total num-
ber of granule cells was not different among different ages in control rats,
there was a progressive decline of total granule cell number with increasing
age after prenatal stress. Interestingly, this decline in prenatally stressed rats
was observable from 3 months of age continuing until 22 months of age.
*
, P
0.05 in comparison to age-matched control group.
Fig. 4. Effect of prenatal stress on number of pyknotic cells in 3-month-,
10-month-, and 22-month-old male rats. With increasing age, there was a
progressive increase in the total number of pyknotic cells. There was no effect
of prenatal stress.
Lemaire et al. PNAS
September 26, 2000
vol. 97
no. 20
11035
NEUROBIOLOGY
BrdUrd-IR cells. The number of BrdUrd-IR cells in prenatally
stressed rats was identical for manipulated and trained animals.
Furthermore, training did not influence the total number of cells
in control [H(1, 10) 1.84, n.s.] and prenatally stressed [H(1,
10) 0.53, n.s., data not shown] rats.
Discussion
The aims of the present study were to investigate (i) whether
prenatal events alter hippocampal plasticity, (ii) to which extent
these possible alterations may undermine behavioral perfor-
mances in spatial memory tasks depending on the hippocampal
formation, and (iii) whether neurogenesis in the dentate gyrus
and learning process can be related. The results show that aging
is accompanied by a progressive decline in cell proliferation. In
addition, prenatally stressed offspring exhibited a greater reduc-
tion in hippocampal cell proliferation by approximately 45% in
all age groups tested. These animals also displayed delayed
learning in a spatial memory task. Furthermore, learning a
spatial memory task stimulated cell proliferation in the dentate
gyrus of control rats whereas prenatal stress altered the ability
of progenitor cells to divide in response to a learning situation.
Prenatal stress induced structural abnormalities in the hippocam-
pal formation. Our results show for the first time that prenatal stress
affects cell proliferation from adolescence until aging. With in-
creasing age, there was a decline in cell proliferation in the dentate
gyrus of control male rats, an effect consistent with previous studies
(24, 25). Prenatally stressed animals displayed a greater reduction
in cell proliferation during aging, suggesting that the early stressful
experience accelerated the age-related decline in hippocampal
plasticity. It remains to be determined whether the age-related
decline in neurogenesis and the effects of prenatal stress are caused
by an alteration in total number of stem cells (or progenitors cells)
or differences in cell cycle length. Interestingly, these effects were
specific to neurogenesis because pyknosis was not modified by
prenatal stress at all ages tested. Because glucocorticoids inhibit
hippocampal cell proliferation (14–16), the increased HPA axis
activity of prenatally stressed animals (1, 3, 5), which is confirmed
here by increased adrenal mass, could explain their reduced neu-
rogenesis. The toxic effects of prolonged secretion of corticosterone
have been extensively studied on hippocampal pyramidal cells (26,
27) but it is not known whether the age-related increase in pyknosis
within the dentate gyrus is related to the activity of the HPA axis.
The lack of effect of prenatal stress on pyknosis suggests the
existence of a complex relationship between corticosterone and cell
death. The maintenance of the effect of prenatal stress on neuro-
genesis during aging can thus be related to the ‘‘glucocorticoid
cascade’’ hypothesis, which proposes that stressful experiences,
through an excessive release of corticosterone, are responsible for
alterations in the structure and function of the hippocampal for-
mation in senescent animals (28).
The effect of aging on total cell number was complex. Indeed,
a decline in cell number was observed between 3 and 10 months
of age but this tendency was reversed later in life. The lack of an
overall effect of aging on granule neuron number was not
consistent with the fact that a net decline in neurogenesis and an
increase in cell death was observed in the same animals. There
is conflicting evidence on the effects of aging on the number of
granule neurons, and the reasons for which variations in this
number may or may not have occurred remain unknown (29–32).
In prenatally stressed rats, despite the early impact of the
manipulation on neurogenesis, an alteration of the total granule
cell number could be observed beginning only at 3 months of age.
This delayed decline in total granule cell number could be
related to the fact that the percentage of granule neurons
produced in adulthood is relatively small as compared with the
1 or 2 millions mature granule neurons. Thus, it is reasonable to
suppose that a relatively long period of neurogenesis reduction
is necessary to affect the total granule cell number and that the
deficits in newborn cells between the beginning of life and 28
postnatal days is negligible as compared with the total pool of
granule neurons. Later in life, deficits in total granule cell
number appear in prenatally stressed rats; this result may be a
consequence of an alteration in cell proliferation, a hypothesis
reinforced by the lack of influence of prenatal stress on pyknosis.
It has been shown that prenatal stress decreased synaptic density
in the CA3 area, the projection site of the granule neurons (33)
and hippocampal density of nitric oxide-producing neurons (34).
Taken together, these data indicate that early experiences alter
Fig. 5. Spatial learning in a water maze. The latency to find the platform by
the control group was significantly less than that observed for the prenatally
stressed group. (Inset) The means over the 5 days of testing.
**
, P 0.01 in
comparison to control group.
Fig. 6. Effect of training in the water maze on cell proliferation in control
and prenatally stressed rats. Acquisition of a spatial task increased the rate of
hippocampal neurogenesis in control rats but not in prenatally stressed rats.
Prenatal stress decreased the total number of BrdUrd-IR cells and, in this
group, the number of newly born cells was identical for manipulated as well
as for trained animals. (Inset) Correlation between cell proliferation and mean
latency to reach the platform.
*
, P 0.05 as compared with manipulated
control group; °, P 0.05 and °°, P 0.01 as compared with control group.
Table 1. Effect of prenatal stress on adrenal weight
Group 28 Days 3 Months 10 Months
Control 260.49 7.74 105.41 8.51 64.66 1.49
Prenatal stress 297.88 12.91* 159.36 6.10** 101.00 14.24*
Data are expressed as a ratio of body weight (mgkg of body weight).
*
, P
0.05 and
**
, P 0.01 as compared to controls.
11036
www.pnas.org Lemaire et al.
the structure of the hippocampal formation. This finding is of
particular interest because the dentate gyrus is associated with
spatial learning and memory (35, 36), which clearly are affected
in prenatally stressed rats (present data and refs. 2, 4, 5).
We show that training in the water maze enhances cell
proliferation in control rats. Previous studies have reported that
survival of newly born cells is enhanced by exposure to an
enriched environment, which also increased spatial learning in
the water maze (37), and by exposure to training in the water
maze (8). So far, the only condition that seems to increase cell
proliferation is voluntary exercise on a running wheel (38). We
show here that training in the water maze enhances cell prolif-
eration in control rats; however, this effect was not seen in a
previous study (38). This discrepancy could be related to the
timing of BrdUrd injections. In this experiment, BrdUrd was
injected at the end of the learning phase, when control rats begin
to reach asymptotic level of performance, instead of throughout
testing. These days were chosen because they correspond to a
period of learning consolidation in which the hippocampal
formation is activated (39). The stimulatory effect of training on
cell proliferation observed in control rats is not caused by an
increased motor activity because these animals cover a shorter
distance than prenatally stressed rats do. Training by itself is not
a sufficient condition to increase cell proliferation because no
such stimulatory effect of training was observed in prenatally
stressed rats. Thus, low cell proliferation is associated with poor
behavioral performance whereas high cell proliferation is asso-
ciated with good learning capabilities. Taken together with the
decreased cell proliferation observed after prenatal stress, the
behavioral deficits induced by prenatal stress suggest an enabling
role for granular cell proliferation in the dentate gyrus to
facilitate spatial memory performances. These results confirm
the importance of the network into which these cells are inserted,
because newborn neurons in the granule cell layer establish
connections with the CA3 pyramidal neurons (10), which are
known to be implicated in spatial memory (35, 36). Reciprocally,
information processing influences brain structures, and spatial-
learning-induced hippocampal plasticity takes place in the GCL-
CA3 network.
The effect of prenatal stress on spatial learning may be the
consequence of a deficit in neurogenesis, which can itself result
from the dysfunction of the HPA axis. Indeed, cognition is
modulated by corticosterone in a complex way (40), and high
levels of corticosterone impair learning and memory (27, 41).
Exposure to the water maze increases corticosterone secretion
(42) and prenatally stressed animals show a delayed habituation
of the corticosterone response to repeated exposure to stress
(43). Thus, it seems reasonable to hypothesize that the prenatal
stress-induced cognitive impairments may result from a pro-
longed corticosterone secretion that inhibits cell proliferation.
Taken together, the results of this study allow us to propose a
pathophysiological path where the effect of prenatal stress on
spatial learning may be the consequence of a deficit in neuro-
genesis that could results from HPA axis dysfunction. Although
a direct effect of corticosterone on neurogenesis is possible,
another hypothesis is that corticosterone acts through serotonin
to reduce neurogenesis. Indeed, serotonin stimulates neurogen-
esis (44), but serotonin levels are reduced by corticosterone (45).
Thus, the decreased serotonin levels observed in prenatally
stressed rats (33) provide further evidence that corticosterone
could be at the origin of the effect of prenatal stress that we
report here. It can be hypothesized that the present results
obtained following maternal restraint stress could be generalized
to other procedures. Indeed, other types of maternal gestational
stress were shown to have similar effects on endocrine and
behavioral parameters (1).
In conclusion, the major impact of our data lies in the
demonstration that deleterious environmental conditions occur-
ring early in life have profound effects on neurogenesis in the
dentate gyrus, an index of hippocampal plasticity, and that these
alterations are associated with impaired performances in a
spatial memory task. Neurogenesis, cell number, and cognitive
capabilities are altered from adulthood to senescence in prena-
tally stressed rats. The fact that stressful experiences during
development could have a long-term effect on hippocampal
function by directly altering its structure recently has been
suggested by Gould and Tanapat (17). Thus, it may be hypoth-
esized that elevated corticosterone levels throughout the lifespan
of these animals could lead to cognitive deficits through devel-
opmental inhibition of neurogenesis (27). Our results reinforce
the hypothesis that many psychopathological affections have
their origin in early developmental influences. More generally,
they show the heuristic value of accurate animal models to better
understand the mechanisms by which early stress and epigenetic
risk factors promote learning disabilities in children and in
age-related disorders, such as Alzheimer’s disease (46).
We thank Drs.G. Rougon, W. Mayo, and P. V. Piazza for helpful comments,
and Dr. M.-F. Montaron and Mrs. C. Brechenmacher for their help with the
confocal analysis. The technical help of Mrs. M. C. Donat, Mrs. J. M.
Claustrat, and O. George is acknowledged. Supported by Institut National
de la Sante´ et de la Recherche Me´dicale, Centre National de la Recherche
Scientifique (98.72.017), and University of Bordeaux II.
1. Weinstok, M. (1997) Neurosci. Biobehav. Rev. 21, 1–10.
2. Szuran, T., Zimmerman, E. & Welzl, H. (1994) Behav. Brain Res. 65, 153–155.
3. Henry, C., Kabbaj, M., Simon, H., Le Moal, M. & Maccari, S. (1994) J. Neuroendocrinol. 6,
341–345.
4. Lordi, B., Poroais, P., Mellier, D. & Caston, J. (1997) Physiol. Behav. 62, 1087–1092.
5. Valle´e, M., Maccari, S., Dellu, F., Le Moal, M., Simon, H. & Mayo, W. (1999) Eur.
J. Neurosci. 11, 2906–2916.
6. Squire, L. R. (1992) Psychol. Rev. 99, 195–231.
7. Altman, J. & Das, G. D. (1965) J. Comp. Neurol. 124, 319–336.
8. Gould, E., Beylin, A., Tanapat, P., Reeves, A. &Shors, T. J. (1999) Nat. Neurosci. 2, 260–265.
9. Gould, E., Tanapat, P., Hastings, N. B. & Shors, J. T. (1999) Trends Cognit. Sci. 3, 186–191.
10. Stanfield, B. B. & Trice, J. E. (1988) Exp. Brain Res. 72, 399406.
11. Cameron, H. A., Wooley, C. S., McEwen, B. S. & Gould, E. (1993) Neuroscience 61, 203–209.
12. Schwegler, H. & Crusio, W. E. (1995) Behav. Brain Res. 67, 2941.
13. Scho¨pke, R., Wolfer, D. P., Lipp, H. P. & Leisinger-Trigona, M. C. (1991) Hippocampus 1,
315–328.
14. Gould, E., Cameron, H. A., Daniels, D. C., Woolley, C. S. & McEwen, B. S. (1992) J. Neuroci.
12, 3642–3650.
15. Rodriguez, J. J., Montaron, M. F., Petry, K. G., Aurousseau, C., Marinelli, M., Premier, S.,
Rougon, G., Le Moal, M. & Abrous D. N. (1998) Eur. J. Neurosci. 10, 2994–3006.
16. Montaron, M. F., Petry, K. G., Rodriguez, J. J., Marinelli, M., Aurousseau, C., Rougon, G.,
Le Moal, M. & Abrous, D. N. (1999) Eur. J. Neurosci. 11, 1479–1485.
17. Gould, E. & Tanapat, P. (1999) Biol. Psychiatry 46, 1472–1479.
18. Chapman, R. H. & Stern, J. (1978) J. Comp. Physiol. Psychol. 92, 1074–1083.
19. Akana, S. F., Shinsako, J. & Dallman, M. F. (1983) Endocrinology 113, 2232–2237.
20. Lemaire, V., Taylor, G. T. & Morme`de, P. (1997) Psychoneuroendocrinology 22, 563–573.
21. Nowakowsky, R. S., Lewin, S. B. & Miller, M. W. (1989) J. Neurocytol. 18, 311–318.
22. Lemaire, V., Aurousseau, C., Le Moal, M. & Abrous, D. N. (1999) Eur. J. Neurosci. 11, 40064014.
23. Howard, C. V. & Reed, M. G. (1998) Unbiased Stereology. Three-Dimensional Measurement
in Microscopy (Springer, New York).
24. Kuhn, H. G., Dickinson-Anson, H. & Gage, F. H. (1996) J. Neurosci. 16, 2027–2033.
25. Seki, T. & Arai, Y. (1995) NeuroReport 6, 2479–2482.
26. Woolley, C., Gould, E. & McEwen, B. S. (1990) Brain Res. 531, 225–231.
27. Sapolsky, R. M., Krey, L. C. & McEwen, B. S. (1985) J. Neurosci. 5, 1222–1227.
28. Sapolsky, R. M. (1992) Stress, the Aging Brain, and the Mechanisms of Neuron Death (MIT
Press, Cambridge, MA).
29. Rapp, P. R. & Gallagher, M. (1996) Proc. Natl. Acad. Sci. USA 93, 9926–9930.
30. Rasmussen, T., Schliemann, T., Sorensen, J. C., Zimmer, J. & West, M. J. (1996) Neurobiol.
Aging 17, 143–147.
31. Landfield, P. W., Baskin, R. & Pitler, T. (1981) Science 214, 5881–5884.
32. Boss, B. D., Peterson, G. M. & Cowan, W. M. (1985) Brain Res. 338, 144–150.
33. Yahashi, A., Nagaoka, M., Yamada, K., Ichitani, Y., Miake, Y. & Okado, N. (1998) Int. J.
Dev. Neurosci. 16, 209–216.
34. Vaid, R. R., Yee, B. K., Shalev, U., Rawlins, J. N., Weiner, I., Feldon, J. & Totterdell, S.
(1997) J. Neurosci. 17, 5599–5609.
35. Jung, M. W. & McNaughton, B. L. (1993) Hippocampus 3, 165–182.
36. McNaughton, B. L., Barnes, C. A., Meltzer, J. & Sutherland, R. J. (1989) Exp. Brain Res. 76, 485–496.
37. Kempermann, G., Kuhn, G. F. & Gage, F. (1997) Nature (London) 386, 493–495.
38. Van Praag, H., Kempermann, G. & Gage, F. H. (1999) Nat. Neurosci. 2, 266–270.
39. Bontempi, B., Laurent-Demir, C., Destrade, C. & Jaffard, R. (1999) Nature (London) 400, 671–675.
40. De kloet, E. R, Oitzl, M. S. & Joe¨ls, M. (1999) Trends Neurosci. 22, 422–426.
41. De Quervain, D. J.-F., Roozendaal, B. & McGaugh, J. L. (1998) Nature (London) 394, 787–790.
42. Sandi, C., Loscertales, M. & Guaza, C. (1997) Eur. J. Neurosci. 9, 637–642.
43. Fride, E., Dan, Y., Feldon, J., Halevy, G. & Weinstock, M. (1986) Physiol. Behav. 37, 681–687.
44. Brezun, J. M. & Daszuta, A. (1999) Neuroscience 89, 999–1002.
45. Chaouloff, F. (1955) Fundam. Clin. Pharmacol. 9, 219–233.
46. Leckman, J. F. (1999) Am. J. Psychiatry 156, 1495–1448.
Lemaire et al. PNAS
September 26, 2000
vol. 97
no. 20
11037
NEUROBIOLOGY
... MS has been found to cause behavioral abnormalities resembling psychotic-like symptoms, such as disruption in the prepulse inhibition response, 11 neuroendocrine alterations associated with stress reactivity, 12,13 and cognitive impairment in juvenile animals. 2,3,14 Various neurotransmitter systems, such as glutamatergic, dopaminergic, and serotonergic activities, can undergo alterations following MS. 13,15−17 Serotonin (5-HT) plays a crucial role in both maturation during sensitive periods and learning-related neuroplasticity, 18 suggesting that inherent differences in the central serotonergic signaling may impact the long-term consequences of ELS. ...
Article
Full-text available
Maternal separation (MS) represents a profound early life stressor with enduring impacts on neuronal development and adult cognitive function in both humans and rodents. MS is associated with persistent dysregulations in neurotransmitter systems, including the serotonin (5-HT) pathway, which is pivotal for mood stabilization and stress-coping mechanisms. Although the novel cannabinoid receptor, GPR55, is recognized for its influence on learning and memory, its implications on the function and synaptic dynamics of 5-HT neurons within the dorsal raphe nucleus (DRN) remain to be elucidated. In this study, we sought to discern the repercussions of GPR55 activation on 5-HT synthesis within the DRN of adult C57BL/6J mice that experienced MS. Concurrently, we analyzed potential alterations in excitatory synaptic transmission, long-term synaptic plasticity, and relevant learning and memory outcomes. Our behavioral assessments indicated a marked amelioration in MS-induced learning and memory deficits following GPR55 activation. In conjunction with this, we noted a substantial decrease in 5-HT levels in the MS model, while GPR55 activation stimulated tryptophan hydroxylase 2 synthesis and fostered the release of 5-HT. Electrophysiological patch-clamp analyses highlighted the ability of GPR55 activation to alleviate MS-induced cognitive deficits by modulating the frequency and magnitude of miniature excitatory postsynaptic currents within the DRN. Notably, this cognitive enhancement was underpinned by the phosphorylation of both NMDA and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. In summary, our findings underscore the capacity of GPR55 to elevate 5-HT synthesis and modify synaptic transmissions within the DRN of juvenile mice, positing GPR55 as a promising therapeutic avenue for ameliorating MS-induced cognitive impairment.
... Accordingly, the resultant smaller hippocampal volume could be a consequence of HL-modulated AHN. In addition, various factors such as environmental enrichment, exercise, stress, and social isolation affect AHN (Gould et al., 1997;Kempermann et al., 1997;Lemaire et al., 2000;van Praag et al., 2000;Kreisel et al., 2014;Voss et al., 2019), suggesting that HL-induced stress and social isolation could indirectly contribute to AHN and hippocampal atrophy. ...
Article
Full-text available
Objectives A growing body of evidence suggests that age-related hearing loss (HL) is associated with morphological changes of the cerebral cortex, but the results have been drawn from a small amount of data in most studies. The aim of this study is to investigate the correlation between HL and gray matter volume (GMV) in a large number of subjects, strictly controlling for an extensive set of possible biases. Methods Medical records of 576 subjects who underwent pure tone audiometry, brain magnetic resonance imaging (MRI), and the Korean Mini-Mental State Exam (K-MMSE) were reviewed. Among them, subjects with normal cognitive function and free of central nervous system disorders or coronary artery disease were included. Outliers were excluded after a sample homogeneity check. In the end, 405 subjects were enrolled. Pure tone hearing thresholds were determined at 0.5, 1, 2, and 4 kHz in the better ear. Enrolled subjects were divided into 3 groups according to pure tone average: normal hearing (NH), mild HL (MHL), and moderate-to-severe HL (MSHL) groups. Using voxel-based morphometry, we evaluated GMV changes that may be associated with HL. Sex, age, total intracranial volume, type of MRI scanner, education level, K-MMSE score, smoking status, and presence of hypertension, diabetes mellitus and dyslipidemia were used as covariates. Results A statistically significant negative correlation between the hearing thresholds and GMV of the hippocampus was elucidated. Additionally, in group comparisons, the left hippocampal GMV of the MSHL group was significantly smaller than that of the NH and MHL groups. Conclusion Based on the negative correlation between hearing thresholds and hippocampal GMV in cognitively normal old adults, the current study indicates that peripheral deafferentation could be a potential contributing factor to hippocampal atrophy.
... Similar to insufficient fetal iron loading, prenatal stress impairs learning [55], increases anxiety and depressive behaviors [56] (reviewed in [10]), and affects dopaminergic and hippocampal development [57,58]. Exposure to stress during prenatal development is linked to cortical thinning and reduced cognitive functioning in offspring [59]. ...
Article
Full-text available
This review presents evidence from animal and human studies demonstrating the possible connection and significant impact of poor iron status and psychological distress on neurocognitive development during pregnancy and the neonatal period, with implications for long-term cognition. Stress and iron deficiency are independently prevalent and thus are frequently comorbid. While iron deficiency and early-life stress independently contribute to long-term neurodevelopmental alterations, their combined effects remain underexplored. Psychological stress responses may engage similar pathways as infectious stress, which alters fundamental iron metabolism processes and cause functional tissue-level iron deficiency. Psychological stress, analogous to but to a lesser degree than infectious stress, activates the hypothalamic–pituitary–adrenocortical (HPA) axis and increases proinflammatory cytokines. Chronic or severe stress is associated with dysregulated HPA axis functioning and a proinflammatory state. This dysregulation may disrupt iron absorption and utilization, likely mediated by the IL-6 activation of hepcidin, a molecule that impedes iron absorption and redistributes total body iron. This narrative review highlights suggestive studies investigating the relationship between psychological stress and iron status and outlines hypothesized mechanistic pathways connecting psychological stress exposure and iron metabolism. We examine findings regarding the overlapping impacts of early stress exposure to iron deficiency and children’s neurocognitive development. We propose that studying the influence of psychological stress on iron metabolism is crucial for comprehending neurocognitive development in children exposed to prenatal and early postnatal stressors and for children at risk of early iron insufficiency. We recommend future directions for dual-exposure studies exploring iron as a potential mediating pathway between early stress and offspring neurodevelopment, offering opportunities for targeted interventions.
Article
Full-text available
Exposure to excess glucocorticoid (GC) during early development is implicated in adult dysfunctions. Reduced adult hippocampal neurogenesis is a well-known consequence of exposure to early life stress or elevated GC, however the effects on neurogenesis during development and effects on other brain regions are not well understood. Using an optogenetic zebrafish model, here we analyse the effects of GC exposure on neurogenesis during development in the whole brain. We identify that the hypothalamus is a highly GC-sensitive region where elevated GC causes precocious development. This is followed by failed maturation and early decline accompanied by impaired feeding, growth, and survival. In GC-exposed animals, the developmental trajectory of hypothalamic progenitor cells is strikingly altered, potentially mediated by direct regulation of transcription factors such as rx3 by GC. Our data provide cellular and molecular level insight into GC-induced alteration of the hypothalamic developmental trajectory, a process crucial for health across the life-course.
Article
Despite major advances, our understanding of the neurobiology of life course socioeconomic conditions is still scarce. This study aimed to provide insight into the pathways linking socioeconomic exposures-household income, last known occupational position, and life course socioeconomic trajectories-with brain microstructure and cognitive performance in middle to late adulthood. We assessed socioeconomic conditions alongside quantitative relaxometry and diffusion-weighted magnetic resonance imaging indicators of brain tissue microstructure and cognitive performance in a sample of community-dwelling men and women (N = 751, aged 50-91 years). We adjusted the applied regression analyses and structural equation models for the linear and nonlinear effects of age, sex, education, cardiovascular risk factors, and the presence of depression, anxiety, and substance use disorders. Individuals from lower-income households showed signs of advanced brain white matter (WM) aging with greater mean diffusivity (MD), lower neurite density, lower myelination, and lower iron content. The association between household income and MD was mediated by neurite density (B = 0.084, p = 0.003) and myelination (B = 0.019, p = 0.009); MD partially mediated the association between household income and cognitive performance (B = 0.017, p < 0.05). Household income moderated the relation between WM microstructure and cognitive performance, such that greater MD, lower myelination, or lower neurite density was only associated with poorer cognitive performance among individuals from lower-income households. Individuals from higher-income households showed preserved cognitive performance even with greater MD, lower myelination, or lower neurite density. These findings provide novel mechanistic insights into the associations between socioeconomic conditions, brain anatomy, and cognitive performance in middle to late adulthood.
Article
Full-text available
Stress during pregnancy has a negative effect on the fetus. However, maternal exercise has a positive effect on the cognitive function of the fetus and alleviates the negative effects of stress. This study aimed to demonstrate whether exercise before pregnancy has a protective effect on prenatal stress-induced impairment of memory, neurogenesis and mitochondrial function in mice offspring. In this experiment, immunohistochemistry, Western blot, measurement of mitochondria oxygen respiration, and behavior tests were performed. Spatial memory and short-term memory of the offspring from the prenatal stress with exercise were increased compared to the offspring from the prenatal stress. The numbers of doublecortin-positive and 5-bromo-2’-deoxyuridine-positive cells in the hippocampal dentate gyrus of the offspring from the prenatal stress with exercise were higher compared to the offspring from the prenatal stress. The expressions of brain-derived neurotrophic factor, postsynaptic density 95 kDa, and synaptophysin in the hippocampus of the offspring from the prenatal stress with exercise were enhanced compared to the offspring from the prenatal stress. Oxygen consumption of the offspring from the prenatal stress with exercise were higher compared to the offspring from the prenatal stress. Exercise before pregnancy alleviated prenatal stress-induced impairment of memory, neurogenesis, and mitochondrial function. Therefore, exercise before pregnancy may have a protective effect against prenatal stress of the offspring.
Article
Full-text available
Accumulating research shows that prenatal exposure to maternal stress increases the risk of behavioral and mental health problems for offspring later in life. However, how prenatal stress affects offspring behavior remains unknown. Here, we found that prenatal stress (PNS) leads to reduced Ahi1, decreased synaptic plasticity and cognitive impairment in offspring. Mechanistically, Ahi1 and GR stabilize each other, inhibit GR nuclear translocation, promote Ahi1 and WDR68 binding, and inhibit DYRK1A and WDR68 binding. When Ahi1 deletion or prenatal stress leads to hyperactivity of the HPA axis, it promotes the release of GC, leading to GR nuclear translocation and Ahi1 degradation, which further inhibits the binding of Ahi1 and WDR68, and promotes the binding of DYRK1A and WDR68, leading to elevated DYRK1A, reduced synaptic plasticity, and cognitive impairment. Interestingly, we identified RU486, an antagonist of GR, which increased Ahi1/GR levels and improved cognitive impairment and synaptic plasticity in PNS offspring. Our study contributes to understanding the signaling mechanisms of prenatal stress-mediated cognitive impairment in offspring.
Article
Full-text available
Exposure to stress can lead to long lasting behavioral and neurobiological consequences, which may enhance the susceptibility for the onset of mental disorders. However, there are significant individual differences in the outcome of stress exposure since only a percentage of exposed individuals may show pathological consequences, whereas others appear to be resilient. In this study, we aimed to characterize the effects of prenatal stress (PNS) exposure in rats at adolescence and to identify subgroup of animals with a differential response to the gestational manipulation. PNS adolescent offspring (regardless of sex) showed impaired emotionality in different pathological domains, such as anhedonia, anxiety, and sociability. However, using cluster analysis of the behavioral data we could identify 70% of PNS-exposed animals as vulnerable (PNS-vul), whereas the remaining 30% were considered resilient (PNS-res). At the molecular level, we found that PNS-res males show a reduced basal activation of the ventral hippocampus whereas other regions, such as amygdala and dorsal hippocampus, show significant PNS-induced changes regardless from vulnerability or resilience. Taken together, our results provide evidence of the variability in the behavioral and neurobiological effects of PNS-exposed offspring at adolescence. While these data may advance our understanding of the association between exposure to stress during gestation and the risk for psychopathology, the investigation of the mechanisms associated to stress vulnerability or resilience may be instrumental to develop novel strategies for therapeutic intervention.
Article
Full-text available
Neuroplasticity refers to the ability of the brain to reorganize and modify its neural connections in response to environmental stimuli, experience, learning, injury, and disease processes. It encompasses a range of mechanisms, including changes in synaptic strength and connectivity, the formation of new synapses, alterations in the structure and function of neurons, and the generation of new neurons. Neuroplasticity plays a crucial role in developing and maintaining brain function, including learning and memory, as well as in recovery from brain injury and adaptation to environmental changes. In this review, we explore the vast potential of neuroplasticity in various aspects of brain function across the lifespan and in the context of disease. Changes in the aging brain and the significance of neuroplasticity in maintaining cognitive function later in life will also be reviewed. Finally, we will discuss common mechanisms associated with age-related neurodegenerative processes (including protein aggregation and accumulation, mitochondrial dysfunction, oxidative stress, and neuroinflammation) and how these processes can be mitigated, at least partially, by non-invasive and non-pharmacologic lifestyle interventions aimed at promoting and harnessing neuroplasticity.
Article
Full-text available
The hippocampus of the rat loses neurons with age, a loss which may eventuate in some of the functional impairments typical of senescence. Cumulative exposure to corticosterone (CORT) over the lifespan may be a cause of this neuronal loss, as it is prevented by adrenalectomy at mid- age. In this study, we demonstrate that prolonged exposure to CORT accelerates the process of cell loss. Rats were injected daily with sufficient CORT to produce prolonged elevations of circulating titers within the high physiological range. Animals treated for 3 months (chronic subjects) resembled aged rats in a number of ways. First, both groups had extensive and persistent depletions of CORT receptors in the hippocampus; in the case of chronic rats, no recovery of receptor concentrations occurred 4 months after the end of steroid treatment. Second, autoradiographic analysis revealed that the receptor depletion was due, in part, to a loss of CORT-concentrating cells, especially in the CA3 cell field. Remaining cells bound significantly less [3H]corticosterone than did those of control rats. Finally, analysis of size distributions of hippocampal cell bodies indicated that chronic subjects lost neurons of the same size as those lost in the aged hippocampus. Furthermore, chronic subjects also had increased numbers of small, darkly staining cells of CA3; these corresponded in size to the dark glia whose numbers increase in the aged hippocampus, and which are thought to infiltrate in response to neuronal damage or destruction. Thus, this study supports the hypothesis that cumulative exposure to CORT over the lifespan may contribute to age-related loss of neurons in the hippocampus, and that prolonged stress or exposure to CORT accelerates this process.
Article
Full-text available
Investigated whether the demasculinizing and feminizing effects of prenatal stress (i.e., stress applied to the mother during pregnancy) in rats reported previously by I. L. Ward (1972) are mediated by activation of the maternal pituitary–adrenal axis. Neither whole-body restraint, with or without hyperthermia, nor ACTH treatment during the last third of gestation had any reliable effect on masculine or feminine sexual behavior in male Sprague-Dawley offspring, although these treatments produced maternal pathology and evidence of maternal adrenocorticoid release. Significant littermate similarity was found for almost every morphological and behavioral measure. Failure to control for the litter variable may account for many previously reported effects of prenatal stress on sexual behavior in rats. The discrepancy between the present and earlier findings is discussed in terms of methodological and theoretical considerations. (36 ref)
Article
Full-text available
The purpose of the present study was to determine whether the demasculinizing and feminizing effects of prenatal stress (i.e., stress applied to the mother during pregnancy) in rats reported previously are mediated by activation of the maternal pituitary-adrenal axis. Neither whole-body restraint, with or without hyperthermia, nor ACTH treatment during the last third of gestation had any reliable effect on masculine or feminine sexual behavior in male Sprague-Dawley offspring, although these treatments produced maternal pathology and evidence of maternal adrenocorticoid release. Significant littermate similarity was found for almost every morphological and behavioral measure. Failure to control for the litter variable may account for many previously reported effects of prenatal stress on sexual behavior in rats. The discrepancy between the present and earlier findings is discussed in terms of methodological and theoretical considerations.
Article
Full-text available
The rat dentate gyrus is unusual among mammalian brain regions in that it shows cell birth well into adulthood. During development, dentate gyrus cell birth is regulated by adrenal steroids. However, it is presently unknown whether cell division in the adult is also mediated by these same factors. In order to determine whether this is the case, we combined adrenalectomy, with or without corticosterone (CORT) replacement, and 3H-thymidine autoradiography, Nissl staining, and immunohistochemistry for the glial cell markers vimentin and glial fibrillary acidic protein (GFAP) as well as for the neuronal marker neuron-specific enolase. Removal of circulating adrenal steroids resulted in a greater density of both GFAP-immunoreactive and vimentin-immunoreactive cells compared to sham-operated animals; CORT replacement prevented increases in both of these cell types. The increase in the density of vimentin-immunoreactive cells probably resulted from an increase in the birth of these cells, as adrenalectomized rats showed greater numbers of 3H-thymidine-labeled vimentin-positive cells compared to sham rats. In contrast, no changes in the number of 3H-thymidine-labeled GFAP-positive cells were observed with adrenalectomy, indicating that the increase in this cell type probably does not involve cell birth. In addition, the density of 3H-thymidine-labeled cells that were not immunoreactive for either glial cell marker and that showed neuronal characteristics was dramatically increased with adrenalectomy. These results suggest that adrenal hormones normally suppress the birth of both glia and neurons in the adult rat dentate gyrus.
Article
We have used Golgi-impregnated tissue to demonstrate that exposure to excess glucocorticoids alters dendritic morphology in a specific population of neurons in the adult rat hippocampus. Daily injection of 10 mg of corticosterone for 21 days resulted in decreased numbers of apical dendritic branch points and decreased total apical dendritic length measured in a 100-μm-thick section in CA3 pyramidal cells compared to sham-injected and non-injected controls. In contrast, no changes were observed in CA3 pyramidal cell basal dendritic morphology. Furthermore, no changes were observed in the dendritic morphology of CA1 pyramidal cells or granule cells of the dentate gyrus. Cross-sectional cell body area of any of the 3 cell types examined in this study was unaffected by corticosterone treatment. Finally, qualitative analysis of Nissl-stained tissue from the same brains revealed increased numbers of darkly staining, apparently shrunken CA3 pyramidal cells in corticosterone treated compared to control brains. The changes in dendritic morphology we have observed may be indicative of neurons in the early stages of degeneration, as prolonged exposure to high levels of corticosterone has been shown by others29 to result in a loss of CA3 pyramidal cells. Additionally, these results suggest possible structural alterations which may occur under physiological conditions in which corticosterone levels are chronically elevated such as in aged animals.
Article
This study investigated the hypotheses that unpredictable prenatal stress (1) has effects on the offspring, similar to those induced by perinatal administration of glucocorticoids and (2) increases the vulnerability to stressful situations at adulthood. Rats were exposed to random noise and light stress throughout pregnancy. Offspring were tested for the development of spontaneous alternation behavior (SA) and at adulthood, their response to novel or aversive situations, open field, extinction and punishment following acquisition of an appetitive response and two-way active avoidance, were assessed. In prenatally stressed rats, the development of SA was significantly delayed. On repeated exposure to an open field they were less active; control rats had elevated plasma corticosterone (CCS) on days 2 and 4 of open field exposure, while prenatally stressed rats had significantly raised plasma CCS after each exposure (days 1–8). Furthermore, punishment-induced suppression of an appetitive response was enhanced. Acquisition of active avoidance was faciliated in female but reduced in male prenatally stressed offspring. It is suggested that random prenatal noise and light stress may cause impairment of development of hippocampal function which lasts into adulthood. This impairment is manifested as an increase in vulnerability and a decrease in habituation to stressful stimuli.
Article
WEINSTOCK, M. Does prenatal stress impair coping and regulation of hypothalamic-pituitary-adrenal axis? NEUROSCI BIOBEHAV REV 21(1),1–10 1997.—Prenatally stressed (PS) human infants and experimental animals show attentional deficits, hyperanxiety and disturbed social behavior. Impaired coping in stressful situations in adult PS monkeys and rodents is associated with dysregulation of the HPA axis, characterized by decreased feedback inhibition of corticotropin-releasing hormone (CRH) and prolonged elevation of plasma glucocorticoids in response to stress. PS rats have higher levels of CRH in the amygdala, fewer hippocampal glucocorticoid receptors and less endogenous opioid and GABA/BDZ (benzodiazepine) inhibitory activity. The mechanisms by which maternal stress hormones induce these long-lasting changes in the developing fetal neuroaxis remain to be elucidated. It is suggested that impaired coping in stressful situations and dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, result from the action of maternal hormones released during stress on the developing fetus. The similarities in coping behavior and dysregulation of the HPA axis in PS animals to those in humans with depression, suggest that gestational stress, at a critical time during fetal development, may increase the propensity to develop this condition. Copyright © 1996 Elsevier Science Ltd.