Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Brain Research Bulletin 71 (2007) 619–627

Diethylstilbestrol alters the population dynamic of neural precursor


cells in the neonatal male rat dentate gyrus
Jorge G. Ramos ∗ , Jorgelina Varayoud, Lucas Monje, Guillermo Moreno-Piovano,
Mónica Muñoz-de-Toro, Enrique H. Luque
Laboratorio de Endocrinologı́a y Tumores Hormonodependientes, School of Biochemistry and Biological Sciences,
Universidad Nacional del Litoral, Casilla de Correo 242, 3000 Santa Fe, Argentina
Received 16 September 2006; received in revised form 21 November 2006; accepted 5 December 2006
Available online 4 January 2007

Abstract
Little is known about how estrogens influence neurogenesis in the newborn male rodent. Herein, we examined the effects of neonatal diethyl-
stilbestrol (DES) exposure on the proliferation and survival of type-1 and type-2 neural precursor cells (NPC) in the dentate gyrus of male rats.
This was achieved by exposing newborn male pups to DES on postnatal day (PND) 1, PND3, PND5, and PND7, sacrificed at PND8 or PND21,
followed by double immunohistochemistry and morphometric analysis of hippocampal dentate gyrus. Furthermore, vascular endothelial growth
factor (VEGF) and brain-derived neurotrophic factor (BDNF) mRNA expression was evaluated in hippocampal tissue blocks by real time RT-PCR.
At PND8, the density of total proliferating NPC decreased in DES-treated animals. This reduction was due to a significant decrease in the mitotic
rate of type-2 cells only, since type-1 NPCs did not show changes in the proliferation index. Type-2 NPCs expressed the cell-cycle inhibitor p27kip1
and its expression was clearly augmented in the DES-treated group. Furthermore, the number of apoptotic cells in the dentate gyrus of DES-treated
rats decreased. Surprisingly, DES treatment enhanced cell survival and increased NPCs proliferation when animals were examined 14 days after
treatment. VEGF mRNA expression showed a positive correlation with NPCs proliferation and BDNF mRNA levels were higher in DES-treated
animals at both time points examined. Collectively, these results indicate that hippocampal NPCs proliferation and survival is a critical target of
DES exposure during the early postnatal period. VEGF and BDNF are proposed as key mediators of DES-induced NPC mitotic response.
© 2006 Elsevier Inc. All rights reserved.

Keywords: Hippocampus; Neural precursors; DES; Proliferation; Apoptosis

1. Introduction and reach the inner most portion of the molecular zone while
type-II NPCs lack these long processes and are characterized
Neurogenesis occurs throughout life in mammals includ- by short cytoplasmic extensions that are tangentially oriented
ing man [14,23]. Neural precursor cells (NPC), located in the to the GCL [16]. Under resting conditions, type-II NPCs are
subgranular zone (SGZ) of the dentate gyrus, proliferate and most actively dividing, and physiologic mitogenic stimuli seem
integrate in hippocampal circuits and may be involved in certain to affect primarily these early types of precursors. Type-I cells
forms of hippocampal-dependent learning [19,47]. Two NPC rarely divide, and their proliferation is not induced by the exper-
subtypes (type-I and type-II NPCs) have been characterized imental manipulations studied so far [13,26].
previously using transgenic mice expressing green fluorescent Previous studies have identified endocrine, neural, and
protein (GFP) under the promoter for nestin, an intermediate fil- experiential factors that regulate the production and survival
ament present in progenitor cells [16]. The two cellular subtypes of late-generated neurons in the rat hippocampus [29,35].
express nestin and can be distinguished using morphological cri- These include negative modulators, such as glucocorticoids
teria [13,16]. Nestin expressing type-I NPCs have long processes and interleukin-6 [34,51], and positive modulators, such as
that go through the granule cell layer (GCL) of the dentate gyrus serotonin, N-methyl-aspartate receptor antagonists and dehy-
droepiandrosterone [1,24].
Estrogens have been shown as regulators of synaptic plas-
∗ Corresponding author. Tel.: +54 342 4575207; fax: +54 342 4575207. ticity, cognitive function, memory, mood and behavior [32];
E-mail address: gramos@fbcb.unl.edu.ar (J.G. Ramos). however, their roles in the control of neuronal and glial prolif-

0361-9230/$ – see front matter © 2006 Elsevier Inc. All rights reserved.
doi:10.1016/j.brainresbull.2006.12.004
620 J.G. Ramos et al. / Brain Research Bulletin 71 (2007) 619–627

eration and differentiation are still not fully understood. Recent 2. Materials and methods
reports have shown that estradiol is able to influence cell pro-
duction in the adult hippocampal formation [41,50]. A single 2.1. Animal treatments
dose of estradiol (10 ␮g/rat) has been shown to be capable
Pups were obtained from timed-pregnant Wistar rats housed under a con-
of increasing NPC proliferation in the SGZ of ovariectomized trolled environment (22 ± 2 ◦ C; lights on from 06:00 to 20:00 h) with free access
adult female rats [49]. In contrast, other studies have demon- to pellet laboratory chow (Cooperación, Buenos Aires, Argentina) and tap water
strated that the density of new cells is negatively correlated supplied from glass bottles. All rats were handled in accordance with the prin-
with plasma estradiol levels in wild adult female meadow voles ciples and procedures outlined in the Guide for the Care and Use of Laboratory
Animals issued by the National Academy of Sciences (USA). All protocols
and laboratory rats [37,38]. This apparent discrepancy could
were approved by the Bioethical Commission of the School of Biochemistry
be explained by a time- and dose-dependent action of estro- and Biological Science of the Universidad Nacional del Litoral.
gen on NPC proliferation [37,38,50]. Moreover, the positive or Seven pregnant dams per treatment group were used to collect offspring for
negative action of estrogens on hippocampal cell proliferation each time point (see below). The day of birth was designated as postnatal day
during adulthood could be mediated by adrenal steroids and their 0 (PND0). Pups were sexed according to ano-genital distance, and each litter
was culled randomly on PND1 to a maximum of 10 neonates, with up to 10
receptors [38].
male pups per litter if possible. When fewer than 10 males were available, an
Some efforts have been taken to elucidate possible mecha- appropriate number of females were retained. All male pups were assigned to
nisms and molecular mediators underlying NPC proliferation one of two groups, and were given sc injections of either DES (0.2 ␮g/pup,
and differentiation. Vascular endothelial growth factor (VEGF) Sigma, St. Louis, MO) dissolved in corn oil or an equal volume (25 ␮l) of corn
has been proposed as a key molecule that could regulate neuro- oil alone on PND1, PND3, PND5, and PND7.
The DES dose used here did not produce signs of acute or chronic toxicity
genesis, mediating the effects of environment on hippocampal
and no significant differences in weight gain between DES-exposed and control
plasticity during adulthood [3]. Most research on the effects of pups were recorded during the experiment (data not shown). On PND8, a set
estrogens on neurogenesis has been performed on adult female of males of both groups was injected i.p. with the thymidine analog bromod-
rodents, and thus, little is known about the consequences of eoxyuridine (BrdU, Sigma, 60 mg/kg of body weight). To determine the NPC
estrogen exposure on hippocampal NPCs proliferation in the proliferation index on PND8, some animals (n = 12 for each group) were killed
by decapitation 4 h after BrdU injection. To evaluate the NPC survival index,
neonatal male brain. There is a great body of evidence prov-
others were sacrificed 14 days (PND21) after BrdU treatment (n = 10 for each
ing that the perinatal period is critical on hippocampal circuitry group). To assess NPC proliferation 2 weeks after DES treatment, another set of
formation [27]. During development, estrogen treatment has rats was injected with BrdU on PND21 and sacrificed 4 h later (n = 13 for each
been shown to affect the volume of CA1 and CA3 hippocam- group). For immunohistochemical procedures, brains from males sacrificed at
pal regions, spatial navigation behavior [21], the strength of each time point were fixed by immersion in 4% paraformaldehyde in PBS for 24 h
at 4 ◦ C. Fixed tissue was dehydrated in an ascending series of ethanol, cleared in
hippocampal long-term potentiation [8,57], and hippocampal-
xylene and embedded in paraffin. For real time RT-PCR analysis, a second set of
dependent learning [30]. The GCL of the hippocampal dentate brains (n = 10 per treatment group in each time point) was quickly removed, and
gyrus has been characterized as a region of continuous but age- the dorsal pole of the hippocampus (including the dentate gyrus) was microdis-
dependent proliferation of NPC in rodents and humans [14,56]. sected, using needles of adequate internal diameters [39]. Tissue blocks were
DNA labeling studies performed in rats have demonstrated that flash frozen in liquid nitrogen and stored at −80 ◦ C until RNA extraction.
NPCs proliferation is intense during the first postnatal week
2.2. Immunohistochemistry
in the SGZ of the dentate gyrus, declining from the second
postnatal week to adulthood [18,44]. Brain serial sections (5 ␮m in thickness) were mounted on 3-aminopropyl
DES is a synthetic estrogen that is frequently used in neona- triethoxysilane (Sigma) coated slides and dried for 24 h at 37 ◦ C. BrdU incor-
tal estrogenization experiments because it is not bound by poration into cells in the S phase of the cell cycle was evaluated as previously
alpha-fetoprotein and penetrate the blood brain barrier [12,46]. described [25]. All primary antibodies were incubated overnight at 4 ◦ C. For
BrdU labeling, a mouse monoclonal antibody against BrdU (clone 85-2C8,
Previously, it has been shown that DES exposure during the Novocastra, Newcastle upon Tyne, UK) was used at a dilution of 1:100.
developmental stage resulted in a marked influence on synapto- Immunostaining of estrogen receptor ␣ (ER␣) was performed using a mouse
genesis and neuronal vulnerability [45]. Several reports have monoclonal antibody raised to full-length recombinant human ER␣ (clone
indicated that neonatal xenoestrogen exposure inhibits sex- 6F11, Novocastra) at a dilution of 1:200, and for the detection of p27kip1 ,
ual differentiation of non-reproductive behaviors, including a polyclonal antibody diluted 1:500 raised against a peptide mapping at the
carboxy terminus of p27 of human origin was evaluated (sc-528, Santa Cruz
hippocampal-dependent tasks, like maze learning and spatial Biotechnology Inc., CA). Reactions were developed using a streptavidin–biotin
location [4,15]. However, very little is known about the effects peroxidase method and diaminobenzidine (DAB) (Sigma) as a chromogen sub-
and mechanisms underlying DES exposure during the early strate. Samples were counterstained with Harris hematoxylin (Biopur, Rosario,
postnatal period on the male hippocampal NPC population Argentina) and mounted with permanent mounting medium (PMyR, Buenos
dynamic. Aires, Argentina). Each immunohistochemical run included positive and nega-
tive controls. For negative controls, the primary antibodies were replaced with
Taking into account the above-mentioned findings, the goal non-immune mouse or rabbit serum (Sigma), or immunohistochemistry (IHC)
of the present study was to determine the effects of DES expo- was performed in samples from animals that did not receive BrdU.
sure on the proliferation, apoptosis and survival of hippocampal
NPC in vivo, during the first week of male rat postnatal life. 2.3. Double IHC
To evaluate the possible molecular mediators of these effects, a
To identify the immunophenotype of the proliferating or quiescent cells, a
real time RT-PCR approach was optimized to quantify VEGF second round of immunolabeling was performed after BrdU or p27kip1 detection
and BDNF mRNA expression levels in the dorsal dentate gyri using anti-nestin (identifying type-1 and type-2 NPCs) [16], anti-calbindin (to
of neonatally estrogenized male rats. detect mature granule neurons), or anti-GFAP (to detect astrocytes) as a second
J.G. Ramos et al. / Brain Research Bulletin 71 (2007) 619–627 621

primary antibody. The sections were incubated overnight at 4 ◦ C with anti-nestin MD). To calculate the total volumes of the portions of the GCL and SGZ ana-
(clone rat 401, BD Pharmingen, San Jose, CA) 1:200, or anti-calbindin-D- lyzed, two consecutive sections every 120 ␮m were selected and the outlines of
28-K (KD-15, Sigma) 1:200, or anti-GFAP (Sigma) 1:100. Reactions were the GCL and the SGZ were drawn with a low-power objective lens (10×). The
developed using a streptavidin–biotin peroxidase method. Visualization of this estimation of GCL and SGZ volumes were made according to the principle of
second round of antigens was achieved by the DAB-nickel intensified technique. Cavalieri [53]. The areas were calculated by means of Image Pro-Plus software
The DAB-nickel solution (2.3 mg DAB, 4 ml 0.05 M Tris–HCl buffer [pH 7.5], and the total volume for each hemisphere was estimated as the sum of the areas
15 ␮l 30% H2 O2 , and 460 ␮l 1% nickel chloride) was added to the samples. multiplied by the distance between the analyzed sections.
After 10 min at 20 ◦ C, the samples were rinsed in running water. Slides were
counterstained with Harris hematoxylin (Biopur) and mounted with permanent 2.6. Real-time PCR analysis
mounting medium.
An optimized RT-PCR protocol was employed to analyze the relative expres-
2.4. In situ detection of apoptosis sion levels of VEGF and BDNF mRNAs in the dentate gyrus of male rats on
PND8 and PND21. Individual samples of hippocampal dentate gyri of each
experimental group were homogenized in TRIzol (Invitrogen, Carlsbad, CA),
Sections were analyzed for in situ detection of cells with DNA strand breaks
and RNA was prepared according to the manufacturer’s protocol. Equal quan-
(apoptotic cells) using the TUNEL technique (ApopTag, Intergen Co., Pur-
tities (4 ␮g) of total RNA were reverse-transcribed into cDNA with AMV
chase, NY). In brief, after deparaffinization and rehydration, sections were
reverse transcriptase (12.5 U; Promega, Madison, WI) using 200 pmol of random
incubated with proteinase K (20 ␮g/ml) (Intergen) for 15 min at 37 ◦ C and then
primers (Promega). Twenty units of ribonuclease inhibitor (RNAout) (Invitro-
treated with hydrogen peroxide in PBS for 10 min at RT to quench endogenous
gen Argentina, Buenos Aires, Argentina) and 100 nmol of a deoxynucleotide
peroxidase activity. Sections were then incubated with a mixture containing
triphosphate mixture were added to each reaction tube in a final volume of 30 ␮l
digoxigenin-labeled deoxynucleotide triphosphate, unlabeled deoxynucleotide
of 1× AMV-RT buffer. Reverse transcription was performed at 42 ◦ C for 90 min.
triphosphate mix, and TdT enzyme in a humidified chamber at 37 ◦ C for 1 h.
Reactions were stopped by heating at 97 ◦ C for 5 min and cooling on ice, fol-
Slides were subsequently rinsed with PBS and incubated with anti-digoxigenin-
lowed by dilution of the reverse-transcribed cDNA with RNAse free water to a
peroxidase for 30 min at 20 ◦ C and substrate–chromogen mixture (DAB, Sigma)
final volume of 60 ␮l. Samples were analyzed in duplicate or triplicate, and a
for 6 min. Samples were counterstained with Mayer hematoxylin, dehydrated,
sample without reverse transcriptase was included in each plate to detect contam-
and mounted with a permanent mounting medium. Negative control slides were
ination by genomic DNA. Primer pairs used for amplification of VEGF, BDNF
ran using the same procedures, except that distilled water was added instead of
and the ribosomal protein L19 cDNAs are shown in Table 1. cDNA levels were
TdT enzyme. As a positive control, involuting rat prostate after the second day
detected using real-time PCR with the DNA Engine Opticon System (Bio-Rad
of castration was processed in an identical manner to the experimental samples.
Laboratories Inc. Waltham, MA) and SYBR Green I dye (Cambrex Corp., East
Rutherford, NJ). For cDNA amplification, 5 ␮l of cDNA were combined with
2.5. Morphometry a mixture containing 2.5 U Taq-DNA polymerase (Invitrogen), 2.5 mM MgCl2
(Invitrogen), 0.2 mM of each of the four dNTPs (Promega), and 10 pmol of each
To avoid interference of possible neurogenetic gradients [9] in differences primer (Invitrogen) in a final volume of 25 ␮l of 1× SYBR Green I PCR Taq
depending on the treatment, the same hippocampal region was analyzed in all buffer. After initial denaturation at 97 ◦ C for 5 min, the reaction mixture was sub-
groups for BrdU and neuronal markers immunohistochemistry and TUNEL jected to successive cycles of denaturation at 96 ◦ C for 45 s, annealing at 60 ◦ C
staining. The area of the hippocampus evaluated in this work was limited to for VEGF and BDNF genes for 30 s, and extension at 72 ◦ C for 1 min. The
the dorsomedial pole of the hippocampus, where the dentate gyrus is horizon- annealing temperature for the amplification of the L19 housekeeping gene was
tally oriented beneath the corpus callosum and the external and internal blades 56 ◦ C. Product purity was confirmed by dissociation curves and random agarose
are joined at the crest (an approximate 0.8 mm block of tissue extending in the gel electrophoresis. No DNA template controls were included in any of the
caudal direction from the first appearance of the GCL) [6,27,40]. The SGZ was assays, yielding no consistent amplification. Calculation of relative expression
defined as a 150 ␮m band immediately adjacent to the hilar surface of the GCL. levels of each target was conducted based on the cycle threshold (CT ) method
The cell counting procedures were performed in the GCL and the SGZ of the [20]. The CT for each sample was calculated using the Opticon Monitor Anal-
dentate gyrus as was previously described [6,40]. To calculate the proportions ysis Software (Bio-Rad Laboratories) with an automatic fluorescence threshold
of each cell phenotype and the density of BrdU-positive cells in the dentate setting. Efficiency of PCR reactions was assessed for each target by amplifica-
gyrus, the total number of BrdU or p27kip1 -labeled cells and their respective tion of serial dilutions (over five orders of magnitude) of cDNA fragments of the
phenotypes were scored in six or more sections of a one-in-six series per ani- transcripts under analysis. Depending on specific PCR conditions, efficiencies
mal. To evaluate the entire dorsomedial pole of the hippocampus the different ranged from 90% to 120%. Accordingly, fold expression over control values was
series of coronal sections were separated in the caudal direction by approxi- calculated for each target by the equation 2−CT , where CT is determined by
mately 120 ␮m. The ER␣(+) cell population and the proportion of apoptotic subtracting the corresponding L19 CT value (internal control) from the specific
cells were evaluated using the same criteria. All data were represented as spatial CT of each target and experimental condition. CT is obtained by subtracting
cell densities (number of cells/mm2 of evaluated area) or relative proportions. the CT of each experimental sample from that of the control sample (taken
All spatial measurements (SGZ and GCL area and volume) were performed as reference value 100). Note that no significant differences in CT values were
using an Olympus BH-2 microscope equipped with a Spot Insight color video observed for L19 between the treatment groups. All PCR products were cloned
camera (Diagnostic Instruments, Sterling Heights, MI) and a digital image analy- using the TA cloning kit (Invitrogen, Argentina) and specificity was confirmed
sis software (Image Pro-Plus 4.1.0.1® system, Media Cybernetics, Silver Spring, by DNA sequencing (data not shown).

Table 1
Primer pairs used in real-time RT-PCR
Gene Primer sequence (5 –3 ) Product size (bp) Genbank accession number
BDNF Forward: AGCCTCCTCTG TCTTTCTGCTGGA 298 NM 012513
Reverse: CTTTTGTCTATGCCCCTGCAGCCTT
VEGF Forward: CTGCTCTCTTGGGTGCACTGG 320 NM 031836.1
Reverse: GGTTTGATCCGCATGATCTGCAT
L19 Forward: GAAATCGCCAATGCCAACTC 290 NM 031103
Reverse: ACCTTCAGGTACAGGCTGTG
622 J.G. Ramos et al. / Brain Research Bulletin 71 (2007) 619–627

2.7. Statistics tate gyrus in controls and DES-treated rats. They were often
organized in clusters, with irregularly shaped nuclei. In the
Values are expressed as mean ± S.E.M.; and comparisons were made using dentate gyrus of control PND8 rats, there were more BrdU-
the Mann–Whitney non-parametric test and the threshold of significance was
fixed at P < 0.05.
positive cells than in age-matched DES-treated rats (Fig. 1,
A versus B). Morphometric analysis demonstrated that treat-
3. Results ment with DES decreased cell proliferation (P < 0.01, Fig. 2A).
As described previously [16], NPCs are characterized by mor-
3.1. Neonatal xenoestrogen exposure alters the NPC phological features and by the expression of the filamentous
population dynamic protein nestin. Nestin-expressing precursor cells fall into two
large categories: type-1 and type-2 NPCs. Type-1 NPC is gener-
3.1.1. NPC proliferation and apoptosis on PND8 ally located in the SGZ and characterized by a long process
Proliferating cells were detected in the molecular layer, (Fig. 1C, inset C1) reaching through the GCL and into the
hilus, SGZ and in the GCL innermost portion of the den- molecular layer of the dentate gyrus. Type-2 NPC lacks long

Fig. 1. Effects of neonatal DES exposure on the NPC population dynamic in male pups at PND8. Male pups were injected from PND1 to PND7 with either vehicle
(A, C, E and G) or 0.2 ␮g of DES (B, D, F and H). BrdU(+) cells were diminished in the dentate gyrus of PND8 DES-treated male rats (A vs. B). The two main
categories of nestin-expressing cells were morphologically characterized in the SGZ: type-1 (C1 inset arrows) and type-2 (C2, inset arrows) NPCs; only type-2 NPC
proliferation index was decreased in DES-treated animals (C vs. D, arrowheads). However, the number of apoptotic cells (E, arrows) was decreased in DES-treated
animals (E vs. F). Expression of p27kip1 was only detected in type-2 NPC (G and H, arrows) and was increased in DES-treated group (G vs. H). GCL, granular cell
layer; SGZ, subgranular zone. Scale bar 100 ␮m.
J.G. Ramos et al. / Brain Research Bulletin 71 (2007) 619–627 623

Fig. 2. Quantification of the results obtained by TUNEL technique and by regular and double immunohistochemistry in the dentate gyrus on PND8. Data are
expressed as spatial cell densities and each column represents the mean ± S.E.M. of atleast four semi-serial sections per animal (n = 12 animals per group). (A) Total
proliferating cells in the SGZ; (B) total NPC proliferation density; (C) NPC types-I and -II proliferation indexes; (D) apoptotic cells; (E) total NPC p27kip1 (+) cells;
(F) NPC type-I and type-II p27kip1 (+) density. Scale bar 100 ␮m.

processes having a round or ovoid nucleus and a soma with 3.1.2. NPC survival on PND21
scant cytoplasm (Fig. 1C, inset C2). The short cytoplasmic To determine if DES treatment affects cell survival, we exam-
extensions of these cells tended to be oriented tangentially ined the number of BrdU(+) cells that remains in the SGZ and
to the GCL. Using double IHC, we detected that type-1 and GCL 14 days after the BrdU injection. At this point, the majority
type-2 cells had incorporated BrdU 4 h after i.p. administra- of the BrdU-labeled cells in both groups was fully incorporated
tion, indicating that both cell types had undergone proliferation into the GCL and they were morphologically similar to the sur-
in controls and in DES-treated males. On PND8, the density rounding mature granule neurons. Surprisingly, the number of
of total BrdU(+)/nestin(+) NPC decreased in DES-treated ani- BrdU(+) cells was higher in the DES-treated rats compared with
mals (P < 0.01, Fig. 1, C versus D and Fig. 2B) however, when controls (P < 0.05, Fig. 3A). Regarding the immunophenotype
BrdU(+)/nestin(+) cells were evaluated identifying each NPC of the surviving BrdU(+) cells, results showed that DES did
type, the decrease in proliferative activity was due to type- not alter the proportion of calbindin (mature neuron type) or
2 cells only (P < 0.01, Fig. 2C). On the same postnatal day, GFAP (mature glial type) positive cells compared with controls
the number of pyknotic and TUNEL(+) cells in the SGZ of (Fig. 3B).
DES-treated rats clearly decreased (P < 0.01, Fig. 1, E ver-
sus F, and Fig. 2D), suggesting an anti-apoptotic effect of the 3.1.3. NPC proliferation on PND21
xenoestrogen exposure during the early postnatal period. In In contrast with the results observed on PND8, DES-treated
accordance with the above-mentioned findings, an increased rats that were injected with BrdU on PND21 showed a sig-
expression of p27kip1 was found in the SGZ of DES pups nificantly higher number of double labeled BrdU(+)/nestin(+)
(Fig. 2E). By double IHC, we could classify which type of cells compared with control rats (P < 0.01, Fig. 4A). In addi-
NPC express p27kip1 , showing that only type-2 NPC expresses tion, the NPC of DES-treated group showed a clearly decreased
this cell-cycle inhibitor and its expression was clearly aug- expression of p27kip1 (P < 0.01, Fig. 4B). At this time point, no
mented in DES-treated group (P < 0.01, Fig. 1, G versus H and differences in the number of apoptotic figures were observed
Fig. 2F). between the controls and DES-treated males (Fig. 4C).
624 J.G. Ramos et al. / Brain Research Bulletin 71 (2007) 619–627

Table 2
Quantitative analysis of GCL and SGZ volumes
Control DES
GCL SGZ GCL SGZ

PND8 0.351 ± 0.081 0.151 ± 0.009 0.339 ± 0.015 0.148 ± 0.008


PND21 0.732 ± 0.021 0.387 ± 0.012 0.799 ± 0.056 0.401 ± 0.022

Results are expressed in mm3 as means ± S.E.M.

3.1.4. ERα expression in NPCs sected dentate gyrus of experimental males on PND8 and
Cells that were immunoreactive for ER␣ were observed in the PND21. Results demonstrated that DES treatment significantly
hilus and the SGZ, and none of them were co-labeled with nestin. decreased the VEGF mRNA expression in the hippocampus of
Finally, no differences were found in the number or distribution PND8 rats (P < 0.01, Fig. 5A). In contrast, 14 days after the
of total ER␣(+) cells in both experimental groups at any time end of the xenoestrogen regime (PND21), VEGF mRNA lev-
point analyzed (data not shown). els were clearly augmented in DES-treated animals (P < 0.01,
Fig. 5A). Additionally, BDNF mRNA levels were higher in
3.1.5. GCL and SGZ volumes DES-treated animals at both times points when compared with
The estimated volumes of the GCL and SGZ analyzed in this control animals (P < 0.01, Fig. 5B).
work are shown in Table 2. No differences were obtained in the
volumetric analysis between DES treated and control animals.

3.2. Hippocampal VEGF and BDNF gene expressions are


affected by neonatal DES treatment

Using quantitative real time RT-PCR we examined the


expression of VEGF and BDNF mRNA in the microdis-

Fig. 4. Effects of neonatal DES exposure on NPC proliferation and apoptosis on


Fig. 3. Two weeks after DES exposure, animals showed a higher number of PND21. BrdU was injected 4 h before sacrifice on PND21. Data are expressed
surviving BrdU(+) cells in the dentate gyrus. (A) Spatial density of BrdU(+) as spatial cell densities and each column represents the mean ± S.E.M. of five
cells and morphological detail of surviving cells in each experimental group semi-serial sections per animal (n = 13 animals per group). (A) PND21 NPC
(arrows) and (B) proportion of BrdU(+) surviving cells that show neuron or proliferation; (B) PND21 NPC p27kip1 (+) density; (C) PND21 apoptotic cells
glial phenotype at PND21. (* P < 0.05 vs. control). density. (* P < 0.05 vs. control).
J.G. Ramos et al. / Brain Research Bulletin 71 (2007) 619–627 625

times and doses. However, little is known about the estrogen


effects on dentate gyrus cell proliferation during the early post-
natal period. Our results show that the DES treatment used was
enough to alter the NPCs population dynamic. At PND8, a clear
down-regulation of type-2 NPC proliferation was observed in
the DES-treated males, suggesting that the early postnatal time
could be a sensitive period of xenoestrogen action on the control
of NPC population dynamic. Moreover, the differences observed
between PND8 versus PND21 and those obtained by others in
adult rats [49,50] suggest that the estrogenic responsiveness of
the NPCs vary at different life-time points. This observation is
important since a growing body of evidence shows that man and
wildlife are exposed to estrogenic active compounds during the
perinatal period [42]. The behavioral and physiological conse-
quences of an estrogenic perturbation on NPC proliferation and
differentiation during early postnatal times are still unknown.
Many reports have shown that perinatal exposure to estrogen-
like compounds alters exploratory and socio-sexual behavior
[17,43]. Future studies could address whether these estrogen-
induced behavioral disorders are related with perturbations in
the proliferation and survival of hippocampal NPCs.
Several reports have been demonstrated the importance of
Fig. 5. Effect of neonatal DES exposure on (A) VEGF mRNA and (B) BDNF a functional septohippocampal pathway for the regulation of
mRNA levels in the rat dentate gyrus on PND8 and PND21. Relative mRNA
levels were measured by real-time RT-PCR and fold expression from control
hippocampal neurogenesis [7,33,52]. There is a growing body
values were calculated for each target by the equation 2−CT . Control values of evidence proving that the cholinergic basal forebrain sys-
were assigned to a reference level of 100 and values are given as mean ± S.E.M. tem plays a survival-promoting role for neuronal progenitors
of atleast three independent determinations. (* P < 0.05 vs. control). and immature neurons within regions of adult neurogenesis
like the SGZ of the dentate gyrus [7]. It has been clearly
4. Discussion shown that presynaptic acetylcholine (Ach) receptors play a cen-
tral role in the modulation of hippocampal Ach release [55].
To our knowledge, this is the first study showing that DES Since the cholinergic forebrain neurons activity is regulated by
exposure of newborn male rats significantly affects the prolifera- estrogens through ER␣ [2] an indirect cholinergic pathway in
tion and survival of NPCs in the hippocampal SGZ. An important DES-mediated control of hippocampal neurogenesis could not
reduction in the proliferative activity of hippocampal NPCs was be ruled out.
observed the day after the end of DES treatment while a signifi- The length of the cell-cycle during neurogenesis depends pri-
cant increase in NPC proliferation was detected in rats sacrificed marily on the duration of the G1 transition and is modulated by
2 weeks after the last DES injection. This biphasic NPC prolif- the enzymatic activity of cyclin-dependent kinases (CDKs) [5].
eration response positively correlated with hippocampal VEGF It has been shown that CDK regulators, like the kip1 protein
mRNA levels, suggests that VEGF could be a key mediator in family, may have a critical role in cell-cycle control of transit
this estrogen-induced event. Taking into account that the prolif- amplifying neural progenitors in the subventricular zone of adult
eration of type-1 NPCs was not affected by the xenoestrogen, it is mice brain [11]. During embryonic development, it has been
tempting to propose that the observed changes in the hippocam- shown that p27kip1 is implicated in the maintenance of differen-
pal cell proliferation were due to DES-induced modifications in tiated neurons in a quiescent state; however, little is known about
type-2 NPCs. Moreover, p27kip1 was only expressed in type-2 the possible roles of this CDK inhibitor during postnatal neu-
cells and was negatively correlated with BrdU immunodetec- rogenesis [5]. Using double IHC together with morphological
tion on PND8 and PND21. Collectively, these results show that criteria, we demonstrated that type-2 NPCs express p27kip1 pro-
type-2 NPC population constitutes a target of DES action. tein and this expression were enhanced with DES treatment. An
Many studies have described that estrogen could affect cell inverse correlation between proliferation and p27kip1 expression
proliferation in the dentate gyrus of adult rats. Recently, Tanapat was observed, suggesting a critical role of this CDK inhibitor in
et al. [50] have shown that an acute treatment with a moderate xenoestrogen-induced quiescent state of type-2 NPCs. Not only
(10 ␮g/rat), but not with a low (1 ␮g/rat) or a high (50 ␮g/rat), a great number of type-2 NPCs remained quiescent in DES-
dose of 17␤-estradiol rapidly increased cell proliferation in treated males on PND8 but a significant decrease in the number
ovariectomized adult animals. Moreover, no differences were of apoptotic cells was also detected at this time point. Both events
observed when the steroid was given during prolonged peri- could contribute to the great proportion of remaining BrdU(+)
ods of time (3 weeks). These results clearly showed that the cells observed on PND21 in the DES group, strongly suggest-
effects of estrogens in the control of cell proliferation on the ing that xenoestrogen exposure enhances the survival of cell
dentate gyrus of adult female rats depended on the selected progeny in the male dentate gyrus, while no effects on NPC
626 J.G. Ramos et al. / Brain Research Bulletin 71 (2007) 619–627

differentiation was detected. Moreover, when BrdU incorpora- versidad Nacional del Litoral (CAI + D 2005 019/118). L.M.
tion was evaluated on PND21, a great number of NPCs were and G.M.P. are fellows of the CONICET and the Universidad
positive in the DES-treated group, suggesting the existence of Nacional del Litoral, respectively. J.V., J.G.R., and E.H.L. are
an internal regulatory mechanism triggered to compensate the career investigators of the CONICET.
initial xenoestrogen-induced declination in NPCs proliferation.
There is a growing body of evidence that suggest that extrin- References
sic agents, such as alcohol or xenoestrogens can alter intrinsic
cellular mechanisms of stem cell fate choices contributing to [1] M. Banasr, M. Hery, J.M. Brezun, A. Daszuta, Serotonin mediates oestro-
altered neurogenesis and/or gliogenesis during central nervous gen stimulation of cell proliferation in the adult dentate gyrus, Eur. J.
Neurosci. 14 (2001) 1417–1424.
system maturation [54]. The compensatory mechanism above- [2] S.H. Bora, Z. Liu, A. Kecojevic, I. Merchenthaler, V.E. Koliatsos, Direct,
mentioned could help to restore the number of required granule complex effects of estrogens on basal forebrain cholinergic neurons, Exp.
cells, diminishing the possible neurological adverse effects of an Neurol. 194 (2005) 506–522.
early estrogenic exposure. Whether this mechanism is inducible [3] L. Cao, X. Jiao, D.S. Zuzga, Y. Liu, D.M. Fong, D. Young, M.J. During,
only at early postnatal times or remains active during adulthood, VEGF links hippocampal activity with neurogenesis, learning and memory,
Nat. Genet. 36 (2004) 827–835.
is under study in our laboratory. [4] R. Carr, F. Bertasi, A. Betancourt, S. Bowers, B.S. Gandy, P. Ryan, S.
VEGF is a hypoxia-inducible protein that promotes angio- Willard, Effect of neonatal rat bisphenol a exposure on performance in the
genesis through receptor tyrosine kinases on endothelial cells. Morris water maze, J. Toxicol. Environ. Health A 66 (2003) 2077–2088.
Recent evidence indicates that VEGF can act as a neurotrophic [5] V.S. Caviness Jr., T. Takahashi, R.S. Nowakowski, The G1 restriction point
factor [31,36] and produces neurogenic effects on NPCs [22]. as critical regulator of neocortical neuronogenesis, Neurochem. Res. 24
(1999) 497–506.
A significant reduction in hippocampal VEGF transcripts was [6] S. Ciaroni, R. Cuppini, T. Cecchini, P. Ferri, P. Ambrogini, C. Cuppini, P.
observed on PND8 in DES-treated rats in parallel with a reduc- Del Grande, Neurogenesis in the adult rat dentate gyrus is enhanced by
tion in NPCs proliferation. On the other hand, on PND21, when vitamin E deficiency, J. Comp. Neurol. 411 (1999) 495–502.
the NPCs proliferation was higher in the estrogenized-rats the [7] C.M. Cooper-Kuhn, J. Winkler, H.G. Kuhn, Decreased neurogenesis after
VEGF mRNA levels were significantly increased. We propose cholinergic forebrain lesion in the adult rat, J. Neurosci. Res. 77 (2004)
155–165.
here that hippocampal VEGF could be a xenoestrogen target [8] D.A. Cordoba Montoya, H.F. Carrer, Estrogen facilitates induction of long
gene and a positive molecular mediator in type-2 NPC prolifer- term potentiation in the hippocampus of awake rats., Brain Res. 778 (1997)
ation. 430–438.
Regarding the role of BDNF, it has been shown that estrogen [9] D. Crespo, B.B. Stanfield, W.M. Cowan, Evidence that late-generated gran-
could up-regulate the BDNF mRNA and protein levels in the rat ule cells do not simply replace earlier formed neurons in the rat dentate
gyrus, Exp. Brain Res. 62 (1986) 541–548.
hippocampus [48]. Our results agree with this study, since we [10] A.M. Davies, The role of neurotrophins in the developing nervous system,
observed a significant increase in BDNF mRNA expression in J. Neurobiol. 25 (1994) 1334–1348.
DES-treated animals. BDNF is emerging as an important media- [11] F. Doetsch, J.M. Verdugo, I. Caille, A. Alvarez-Buylla, M.V. Chao, P.
tor of activity-dependent modifications in synaptic strength [28] Casaccia-Bonnefil, Lack of the cell-cycle inhibitor p27Kip1 results in selec-
and plays an important role in the survival and growth of neu- tive increase of transit-amplifying cells for adult neurogenesis, J. Neurosci.
22 (2002) 2255–2264.
rons [10]. However, the possible relationships between BDNF [12] L.L. DonCarlos, M. McAbee, D.S. Ramer-Quinn, D.M. Stancik, Estrogen
and NPCs proliferation remain unknown. Taking into account receptor mRNA levels in the preoptic area of neonatal rats are responsive
that we observed a decrease in apoptotic levels and an increase to hormone manipulation, Brain Res. Dev. Brain Res. 84 (1995) 253–260.
in new granule neurons survival in DES-treated rats, the hypoth- [13] J.G. Emsley, B.D. Mitchell, G. Kempermann, J.D. Macklis, Adult neuro-
esis that BDNF could be a critical mediator of these events could genesis and repair of the adult CNS with neural progenitors, precursors,
and stem cells, Prog. Neurobiol. 75 (2005) 321–341.
not be ruled out. We also suggest that VEGF is a key factor in [14] P.S. Eriksson, E. Perfilieva, T. Bjork-Eriksson, A.M. Alborn, C. Nordborg,
type-2 NPC regulation of proliferation, and that BDNF could D.A. Peterson, F.H. Gage, Neurogenesis in the adult human hippocampus,
be responsible for extended survival times of the neuronal cell Nat. Med. 4 (1998) 1313–1317.
progeny. More studies are necessary to confirm this hypothesis. [15] F. Farabollini, S. Porrini, S.D. Della, F. Bianchi, F. Dessi-Fulgheri, Effects
of perinatal exposure to bisphenol A on sociosexual behavior of female and
male rats, Environ. Health Perspect. 3 (Suppl. 110) (2002) 409–414.
Conflict of interest [16] V. Filippov, G. Kronenberg, T. Pivneva, K. Reuter, B. Steiner, L.P.
Wang, M. Yamaguchi, H. Kettenmann, G. Kempermann, Subpopulation of
The authors declare that there is no conflict of interest that nestin-expressing progenitor cells in the adult murine hippocampus shows
would prejudice the impartiality of this scientific work. electrophysiological and morphological characteristics of astrocytes, Mol.
Cell. Neurosci. 23 (2003) 373–382.
[17] T. Fujimoto, K. Kubo, S. Aou, Prenatal exposure to bisphenol A impairs
Acknowledgements sexual differentiation of exploratory behavior and increases depression-like
behavior in rats., Brain Res. 1068 (2006) 49–55.
The authors thank Mr. Juan C. Villarreal and Mr. Juan Grant [18] E. Gould, P. Tanapat, Stress and hippocampal neurogenesis, Biol. Psychi-
for their technical assistance and animal care. This study was atry 46 (1999) 1472–1479.
supported by grants from the Argentine National Council for [19] N.B. Hastings, E. Gould, Rapid extension of axons into the CA3 region by
adult-generated granule cells, J. Comp. Neurol. 413 (1999) 146–154.
Science and Technology (CONICET, CIC Grant 652/04), the [20] R. Higuchi, C. Fockler, G. Dollinger, R. Watson, Kinetic PCR analysis:
Argentine National Agency for the Promotion of Science and real-time monitoring of DNA amplification reactions, Biotechnology (NY)
Technology (ANPCyT) (PICT 2003, No. 13-4737) and the Uni- 11 (1993) 1026–1030.
J.G. Ramos et al. / Brain Research Bulletin 71 (2007) 619–627 627

[21] C. Isgor, D.R. Sengelaub, Prenatal gonadal steroids affect adult spatial [38] B.K. Ormerod, T.T. Lee, L.A. Galea, Estradiol initially enhances but sub-
behavior CA1 and CA3 pyramidal cell morphology in rats, Horm. Behav. sequently suppresses (via adrenal steroids) granule cell proliferation in the
34 (1998) 183–198. dentate gyrus of adult female rats, J. Neurobiol. 55 (2003) 247–260.
[22] K. Jin, Y. Zhu, Y. Sun, X.O. Mao, L. Xie, D.A. Greenberg, Vascular [39] M. Palkovits, M.J. Brownstein, Microdissection of brain areas by the punch
endothelial growth factor (VEGF) stimulates neurogenesis in vitro and technique, in: A.C. Cuello (Ed.), Brain Microdissection Techniques Chich-
in vivo, Proc. Natl. Acad. Sci. U.S.A. 99 (2002) 11946–11950. ester, John Wiley and Sons, 1983, pp. 1–36.
[23] M.S. Kaplan, J.W. Hinds, Neurogenesis in the adult rat: electron micro- [40] T.D. Palmer, A.R. Willhoite, F.H. Gage, Vascular niche for adult hippocam-
scopic analysis of light radioautographs, Science 197 (1977) 1092–1094. pal neurogenesis, J. Comp. Neurol. 425 (2000) 479–494.
[24] K.K. Karishma, J. Herbert, Dehydroepiandrosterone (DHEA) stimulates [41] M. Perez-Martin, V. Salazar, C. Castillo, C. Ariznavarreta, I. Azcoitia, L.M.
neurogenesis in the hippocampus of the rat, promotes survival of newly Garcia-Segura, J.A. Tresguerres, Estradiol and soy extract increase the pro-
formed neurons and prevents corticosterone-induced suppression, Eur. J. duction of new cells in the dentate gyrus of old rats, Exp. Gerontol. 40
Neurosci. 16 (2002) 445–453. (2005) 450–453.
[25] L. Kass, J. Varayoud, H. Ortega, d.T. Munoz, E.H. Luque, Detection of [42] M. Pombo, L. Castro-Feijoo, Endocrine disruptors, J. Pediatr. Endocrinol.
bromodeoxyuridine in formalin-fixed tissue DNA denaturation following Metab. 18 (Suppl. 1) (2005) 1145–1155.
microwave or enzymatic digestion pretreatment is required, Eur. J. His- [43] S. Porrini, V. Belloni, S.D. Della, F. Farabollini, G. Giannelli, F. Dessi-
tochem. 44 (2000) 185–191. Fulgheri, Early exposure to a low dose of bisphenol A affects socio-sexual
[26] G. Kronenberg, K. Reuter, B. Steiner, M.D. Brandt, S. Jessberger, M. behavior of juvenile female rats, Brain Res. Bull. 65 (2005) 261–266.
Yamaguchi, G. Kempermann, Subpopulations of proliferating cells of the [44] R.M. Sapolsky, M.J. Meaney, Maturation of the adrenocortical stress
adult hippocampus respond differently to physiologic neurogenic stimuli, response: neuroendocrine control mechanisms and the stress hyporespon-
J. Comp. Neurol. 467 (2003) 455–463. sive period, Brain Res. 396 (1986) 64–76.
[27] H.G. Kuhn, H. Dickinson-Anson, F.H. Gage, Neurogenesis in the den- [45] K. Sato, N. Matsuki, Y. Ohno, K. Nakazawa, Effects of 17beta-estradiol
tate gyrus of the adult rat: age-related decrease of neuronal progenitor and xenoestrogens on the neuronal survival in an organotypic hippocampal
proliferation, J. Neurosci. 16 (1996) 2027–2033. culture, Neuroendocrinology 76 (2002) 223–234.
[28] E.S. Levine, C.F. Dreyfus, I.B. Black, M.R. Plummer, Brain-derived neu- [46] L. Savu, C. Benassayag, G. Vallette, E.A. Nunez, Ligand properties of
rotrophic factor rapidly enhances synaptic transmission in hippocampal diethylstilbestrol: studies with purified native and fatty acid-free rat alpha
neurons via postsynaptic tyrosine kinase receptors, Proc. Natl. Acad. Sci. 1-fetoprotein and albumin, Steroids 34 (1979) 737–748.
U.S.A. 92 (1995) 8074–8077. [47] T.J. Shors, D.A. Townsend, M. Zhao, Y. Kozorovitskiy, E. Gould, Neu-
[29] D.C. Lie, H. Song, S.A. Colamarino, G.L. Ming, F.H. Gage, Neurogenesis rogenesis may relate to some but not all types of hippocampal-dependent
in the adult brain: new strategies for central nervous system diseases, Annu. learning, Hippocampus 12 (2002) 578–584.
Pharmacol. Rev. Toxicol. 44 (2004) 399–421. [48] D.T. Solum, R.J. Handa, Estrogen regulates the development of brain-
[30] V.N. Luine, S.T. Richards, V.Y. Wu, K.D. Beck, Estradiol enhances learning derived neurotrophic factor mRNA and protein in the rat hippocampus,
and memory in a spatial memory task and effects levels of monoaminergic J. Neurosci. 22 (2002) 2650–2659.
neurotransmitters, Horm. Behav. 34 (1998) 149–162. [49] P. Tanapat, N.B. Hastings, A.J. Reeves, E. Gould, Estrogen stimulates a
[31] H. Matsuzaki, M. Tamatani, A. Yamaguchi, K. Namikawa, H. Kiyama, M.P. transient increase in the number of new neurons in the dentate gyrus of the
Vitek, N. Mitsuda, M. Tohyama, Vascular endothelial growth factor rescues adult female rat, J. Neurosci. 19 (1999) 5792–5801.
hippocampal neurons from glutamate-induced toxicity: signal transduction [50] P. Tanapat, N.B. Hastings, E. Gould, Ovarian steroids influence cell pro-
cascades, FASEB J. 15 (2001) 1218–1220. liferation in the dentate gyrus of the adult female rat in a dose- and
[32] B.S. McEwen, S.E. Alves, Estrogen actions in the central nervous system, time-dependent manner, J. Comp. Neurol. 481 (2005) 252–265.
Endocr. Rev. 20 (1999) 279–307. [51] L. Vallieres, I.L. Campbell, F.H. Gage, P.E. Sawchenko, Reduced hip-
[33] P. Mohapel, G. Leanza, M. Kokaia, O. Lindvall, Forebrain acetylcholine pocampal neurogenesis in adult transgenic mice with chronic astrocytic
regulates adult hippocampal neurogenesis and learning, Neurobiol. Aging production of interleukin-6, J. Neurosci. 22 (2002) 486–492.
26 (2005) 939–946. [52] B.K. Van der, J. Mulder, J.N. Keijser, B.J. Eggen, P.G. Luiten, E.A. Van
[34] M.F. Montaron, K.G. Petry, J.J. Rodriguez, M. Marinelli, C. Aurousseau, G. der Zee, Input from the medial septum regulates adult hippocampal neuro-
Rougon, M. Le Moal, D.N. Abrous, Adrenalectomy increases neurogenesis genesis, Brain Res. Bull. 67 (2005) 117–125.
but not PSA-NCAM expression in aged dentate gyrus, Eur. J. Neurosci. 11 [53] H. van Praag, B.R. Christie, T.J. Sejnowski, F.H. Gage, Running enhances
(1999) 1479–1485. neurogenesis, learning, and long-term potentiation in mice, Proc. Natl.
[35] M. Naylor, K.K. Bowen, K.A. Sailor, R.J. Dempsey, R. Vemuganti, Acad. Sci. U.S.A. 96 (1999) 13427–13431.
Preconditioning-induced ischemic tolerance stimulates growth factor [54] M.C. Vemuri, C.S. Chetty, Alcohol impairs astrogliogenesis by stem cells
expression and neurogenesis in adult rat hippocampus, Neurochem. Int. in rodent neurospheres, Neurochem. Int. 47 (2005) 129–135.
47 (2005) 565–572. [55] E.S. Vizi, J.P. Kiss, Neurochemistry and pharmacology of the major hip-
[36] O.O. Ogunshola, A. Antic, M.J. Donoghue, S.Y. Fan, H. Kim, W.B. Stew- pocampal transmitter systems: synaptic and non-synaptic interactions,
art, J.A. Madri, L.R. Ment, Paracrine and autocrine functions of neuronal Hippocampus 8 (1998) 566–607.
vascular endothelial growth factor (VEGF) in the central nervous system, [56] C.G. Wasterlain, F. Plum, Vulnerability of developing rat brain to electro-
J. Biol. Chem. 277 (2002) 11410–11415. convulsive seizures, Arch. Neurol. 29 (1973) 38–45.
[37] B.K. Ormerod, L.A. Galea, Reproductive status influences cell proliferation [57] C.S. Woolley, B.S. McEwen, Estradiol mediates fluctuation in hippocampal
and cell survival in the dentate gyrus of adult female meadow voles: a synapse density during the estrous cycle in the adult rat, J. Neurosci. 12
possible regulatory role for estradiol, Neuroscience 102 (2001) 369–379. (1992) 2549–2554.

You might also like