You are on page 1of 11

BIOLOGY OF REPRODUCTION 63, 559569 (2000)

Expression, Ontogeny, and Regulation of Hypoxia-Inducible Transcription Factors in


the Human Placenta1

Augustine Rajakumar and Kirk P. Conrad2


Departments of Obstetrics, Gynecology and Reproductive Sciences and of Cell Biology and Physiology, University of
Pittsburgh School of Medicine and Magee-Womens Research Institute, Pittsburgh, Pennsylvania 15213

ABSTRACT 1213the latter being not significantly different from val-


ues measured concurrently in the endometrium. The higher
Placental hypoxia likely plays an important role in both nor-
levels at Weeks 1213 of pregnancy presumably result from
mal placental development and pathology. Yet, the molecular
mechanisms of hypoxia signaling in this organ are virtually un-
a rise in intervillous blood flow that, at least in part, stems
explored. Therefore, we investigated the expression of the hyp- from trophoblast invasion and physiological remodeling
oxia inducible transcription factors (HIF) in normal human pla- of uterine spiral arteries from small-resistance caliber ves-
centas spanning the first trimester to term. Several key obser- sels to large-capacitance uteroplacental arteries allowing for
vations emerged: 1) HIF-1a and -2a mRNA were present in pla- unimpeded blood flow into the intervillous space [1]. Thus,
centas of all gestational ages but with greater variability during during the first 10 wk or so of development, the human
early pregnancy; 2) overall, HIF-1a mRNA was expressed at a placenta resides in a relatively hypoxic environment.
constant level in all placentas, whereas HIF-2a mRNA increased The proper and timely proliferation and differentiation
significantly with gestational age; 3) both HIF-1a and -2a pro- of the villous cytotrophoblast stem cell are crucial to ade-
tein decreased significantly with gestational age; and 4) HIF-1a quate placentation [4]. Generally speaking, proliferation of
and -2a immunoreactivity were overlapping in cellular distri- villous cytotrophoblast is required for adequate supply of
bution being expressed by the syncytiotrophoblast, villous cy- these cells. They differentiate into the syncytiotrophoblast,
totrophoblast, and fetoplacental vasculature with both nuclear which lines the villous placenta and separates maternal
and cytoplasmic localization. Next, we studied the regulation of blood from villous core. This syncytial cell layer performs
these transcription factors by oxygen using placental villous ex- numerous functions critical to successful pregnancy out-
plants in culture from first-trimester and term placentas. The come, including gas and nutrient transport and pregnancy
major findings were 1) HIF-1a and -2a protein, but not mRNA, hormone production. Alternatively, some of the villous cy-
was induced by hypoxia in the placental villous explants; 2) HIF- totrophoblast stem cells in the so-called anchoring villi that
1a DNA-binding activity was also stimulated by hypoxia; and 3) abut the maternal decidua invade the uterine parenchyma
glucose transporter-1 mRNA (a known target of HIF) was also
and spiral arteries as extravillous trophoblasts. Cytotropho-
increased by hypoxia in placental villous explants. We suggest
that physiological hypoxia contributes to the increased expres-
blast phenotype is influenced by oxygen; for instance, hyp-
sion of HIF-1a and -2a protein in early placentas and that reg- oxia stimulates the proliferation of villous cytotrophoblast
ulation of these transcription factors by hypoxia in the human cells while impairing their invasion [56]. Successful preg-
placenta occurs at the level of protein and not mRNA. nancy outcome also depends on the proper development of
the fetoplacental vasculature in the villous core, which pro-
developmental biology, gene regulation, placenta, syncytiotro- vides for the delivery of oxygen and nutrients from the
phoblast, trophoblast intervillous space to the fetus [4]. Thus, vasculogenesis and
angiogenesis, as well as the entire repertoire of associated
INTRODUCTION growth factors, are remarkably active during placental de-
Successful pregnancy outcome is based on healthy pla- velopment. These processes are well known to be critically
cental development that involves effective trophoblast an- regulated by oxygen. Finally, local ischemia/hypoxia has
choring to and invasion of the uterus (reviewed in [1]). In been implicated in the abnormal cytotrophoblast differen-
the early stages of human placental development, that is, tiation and placental development in preeclampsia [710].
before the 10th wk of gestation, the blood flow to the in- The molecular consequences of cellular hypoxia have
tervillous space is low [2]. The only available data on pla- been extensively reviewed in recent years (e.g., [11]). A
cental oxygen levels were collected by Rodesch et al. [3], variety of genes are modulated as an adaptive response to
who used a transcervical polarographic oxygen electrode to hypoxia, such as those involved in glucose transport, gly-
measure the partial pressure of oxygen in the intervillous colysis, angiogenesis, and erythropoiesis [11]. A prerequi-
space. They obtained values of 17.9 6 6.9 mmHg at Ges- site for many hypoxia-mediated responses is the basic-he-
tational Weeks 810 and of 60.7 6 8.5 mmHg at Weeks lix-loop-helix (bHLH)-Per-Arnt-Sim (PAS) family member
transcription factors HIF-1a [12] and HIF-2a [13, 14],
1
This work was supported by National Institutes of Health Grants PO1 which heterodimerize with the aryl hydrocarbon nuclear
HD30367, RO1 HL56410, and Research Career Development Award translocator protein (ARNT or HIF-1b; 11, 12) and then
KO4 HD01098. bind to the hypoxia response element (HRE; core consensus
Correspondence: Kirk P. Conrad, Magee-Womens Research Institute, sequence59-CGTG-39) in responsive genes. Although the
2

204 Craft Ave., Pittsburgh, PA 15213. FAX: 412 641 1503; e-mail:
rsikpc@mail.magee.edu
identity of the oxygen sensor upstream of the HIF tran-
scription factors remains elusive [15], the molecular mech-
anisms of HIF activation by hypoxia are becoming clearer
Received: 7 February 2000.
First decision: 9 March 2000.
and include protein stabilization [16, 17], thiol-redox reg-
Accepted: 30 March 2000. ulation [18, 19], and interaction with CBP/p300 [19, 20].
Q 2000 by the Society for the Study of Reproduction, Inc. Interestingly, there are agents besides hypoxia that have
ISSN: 0006-3363. http://www.biolreprod.org been shown to activate HIF under normoxic conditions;
559
560 RAJAKUMAR AND CONRAD

these include insulin and IGF-1 [21, 22] and v-SRC [23], it has been reported in stationary cultures of hepatoma cells
as well as transition metals, such as cobalt chloride, and maintained under normoxic conditions that the formation
iron-chelating agents [24]. Finally, HIF transcription factors of unstirred fluid layers leads to reduced pericellular pO2
have been shown to be critical for mouse development. and consequently, to elevated erythropoietin secretion ([32,
Indeed, HIF-1a [25], -2a [26], and -1b [27] homozygous 33] and unpublished results). After a medium change and
knockout mice all manifested embryonic lethality. the addition of any treatments, the plates were again placed
The contribution of HIF to human placental development on the orbital shaker at 378C for various times under either
is virtually unexplored. In a survey of various tissues in normoxia or reduced O2 (hypoxia; 2% O2-5% CO2-balance
humans, Weiner et al. [28] demonstrated the expression of nitrogen) in the cell culture incubators. When the tissues
both HIF-1a and -1b in the term human placenta by North- were treated with cobalt chloride (final concentration of 100
ern analysis. The presence of HIF-1 DNA binding activity mM), the tissues were incubated under normoxic condi-
in isolated cytotrophoblast cells from term placenta was tions. In experiments involving actinomycin D (10 mg/ml)
documented by electrophoretic mobility shift assay [29]. or cyclohexamide (100 mM) to inhibit transcription and
Also, a survey of transcription factor mRNA expression by translation, respectively, the villous explants were pretreat-
isolated trophoblasts from first-trimester human placenta ed for 2 h and then subjected to either the normoxic or
was recently published and included HIF-1a, -2a, and -1b hypoxic incubation conditions in the continuous presence
[30]. Accordingly, there were three major objectives of the of the inhibitor.
present work: 1) to determine the relative abundance of Using a Tucker chamber and a radiometer O2 electrode
HIF-1a and -2a mRNA and protein in the human placenta [34], the pO2 of the hypoxic cell culture incubator was rou-
from early gestation to term; 2) to investigate the regulation tinely found to be 1520 torr, comparable to the levels ob-
of HIF mRNA-, protein-, and DNA-binding activity by ox- served in the intervillous space during early gestation. To
ygen in the human placenta by using villous explant cul- achieve a more severely hypoxic environment of 1% O2,
tures from first-trimester and term placentas; and 3) to ex- modular incubation chambers (Billups-Rotherberg Inc., Del
amine the cellular localization of HIF-1a and -2a protein Mar, CA) were used. These chambers were filled with a 1%
in the first-trimester and term human placentas. Given the O2-5% CO2-balance nitrogen gas mixture. The pO2 was
major role of oxygen in the regulation of normal and ab- routinely 510 torr, which was well maintained for at least
normal placental development and function, the molecular 24 h. These chambers were then placed in the cell culture
mechanisms of oxygen transduction by this organ require incubator for the 378C environment. Cytotoxicity was mon-
further elucidation. itored for each experiment based on the release of lactate
dehydrogenase into the medium and was consistently found
MATERIALS AND METHODS to be 610%, irrespective of the O2 atmosphere, as previ-
ously reported [31].
Placental Collection and Processing
Placentas were obtained from women with normal preg- Hepatoma G2 Cell Culture
nancies undergoing elective cesarean section at term and Hepatoma G2 (HepG2) cells were obtained from the
from women undergoing elective pregnancy terminations at American Type Culture Collection (Manassas, VA). These
512 gestational wk under approval of the Institutional In- cells were used as a positive control for HIF expression
ternal Review Board of Magee-Womens Hospital. [35]. The cells were seeded at a density of 50 000 per
For evaluating the expression and ontogeny of the hyp- square centimeter and were grown in Eagle minimum es-
oxia-inducible transcription factors (HIF), the placental tis- sential medium supplemented with nonessential amino ac-
sues were snap-frozen or placed into 10% formalin imme- ids (Mediatech, Cellgro, Herndon, VA), sodium pyruvate
diately after extraction from the uterus. For evaluating the (1.0 mM; Gibco-BRL, Grand Island, NY), 10% FBS, and
regulation of HIF expression in placental tissues, villous antibiotics.
explants were prepared as described by Benyo et al. [31],
with modifications. For term placenta, several cotyledons Electrophoretic Mobility Shift Assay
were excised at random and rinsed extensively in sterile
saline to remove blood. Decidua and large vessels were Mobility shift assays were carried out using modified
removed from the villous placenta by blunt dissection. The procedures of Semenza et al. [36]. For the preparation of
villous tissue was then finely dissected into 510 mg pieces nuclear extracts, villous explants were rapidly pooled from
while in the bath of sterile saline. The pieces were exten- four to six wells (about 300 mg total wet weight). The
sively washed two or three more times before use. First- excess medium was blotted on a sterile 4 3 4 gauze, and
trimester villi were prepared in a similar fashion. the tissues were transferred into four volumes of buffer A
(10 mM Tris-HCl, pH 7.8; 1.5 mM MgCl2; and 10 mM
Villous Explant Culture KCl containing 0.1% Triton-X 100). Buffers A, C, and D
used in the nuclear extract preparation contained 0.5 mM
We placed 30 to 50 mg of villous tissue into each well dithiothreitol (DTT), 0.4 mM phenymethylsulfonyl fluo-
of a 24-well plate (Becton Dickinson, Franklin Lakes, NJ) ride, 2 mg per milliliter each of leupeptin, pepstatin, and
containing 1.0 ml of Medium 199 (Mediatech, Cellgro, aprotinin, as well as 1 mM sodium vanadate. Care was
Herndon, VA) supplemented with 10% fetal bovine serum taken to process the tissue as quickly as possible, and all
(FBS; Summit Technology, Ft. Collins, CO) and antibiotics. the steps were carried out at 48C. Villous tissue was ho-
Explants were incubated at 378C for a 1224 h preincuba- mogenized in buffer A using a Tissuemizer (Tekmar, Cin-
tion period on an orbital shaker (60 rpm, Belly Dancer; cinnati, OH) at setting 60 for 45 sec. Cell lysis was mon-
Stovall Life Science Inc., Greensboro, NC) under standard itored microscopically by staining with hematoxylin. The
tissue culture conditions of 5% CO2-balance room air (nor- homogenate was centrifuged at 850 3 g for 5 min to pellet
moxia; 20.94% O2) in a cell culture incubator (Forma Sci- the nuclei. The crude nuclear pellet was resuspended in 2.5
entific, Marietta, OH). Cultures were gently shaken because volumes of buffer C (20 mM Tris-HCl, pH 7.8; 0.42 M
HIF-1a, -2a, AND -1b IN THE HUMAN PLACENTA 561

NaCl; 1.5 mM MgCl2; and 20% glycerol). When preparing of RNAwiz using a Tissuemizer. After preparation, the
nuclear extracts from HepG2 cells, we substituted 0.42 M RNA pellet was dissolved in a minimal volume of RNase-
KCl for the NaCl. The nuclear proteins were extracted by free water or in RNase-free water containing 0.5% SDS.
gentle rocking at 48C for 30 min. The extract was centri- The amount of total RNA was estimated by spectrophotom-
fuged at 21 000 3 g for 30 min and then dialyzed against etry (A260). One optical density was taken to be 40 mg/ml.
200 volumes of buffer D (20 mM Tris-HCl, pH 7.8, 0.1 M Ten to 20 mg of total RNA was separated on formaldehyde
KCl, 0.2 mM EDTA, 20% glycerol) for 4 h. The dialyzed containing 1.5% agarose gels and transferred to nylon
nuclear extract was subjected to mild sonication with a mi- membranes (MSI, Westborough, MA). RNA on the mem-
crotip for 5 sec (Sonifier-Cell Disruptor; Heat Systems-Ul- branes was cross-linked by using a Biotech UV-cross-linker
trasonics Inc., Plainview, NY) and then was centrifuged at (Fisher).
21 000 3 g for 15 min. The cleared supernatant was stored For the preparation of the cDNA probes, plasmids pBS/
in aliquots at 2708C until further use. Protein concentration HIF-1a3.23T7 (EcoRI digest of the complete cDNA pro-
was estimated by the Bradford method using reagents from duces three bands of sizes 2063, 1011, and 604 base pairs
Bio-Rad (Hercules, CA). [bp]), and plasmid pGEX2T/ARNT-C (HIF-1b, an 800-bp
For the preparation of the probe, oligonucleotide repre- EcoRI/BamHI fragment) were generously provided by Dr.
senting the erythropoietin gene HRE sequence 5 9 - Gregg Semenza. Plasmid hEPAS-pcDNA3 (HIF-2a, a
GCCCTACGTGCTGTCTCA (200 ng) was end labeled us- 2818-bp BamHI fragment) was a kind gift from Dr. Steven
ing 50 mCi g32P-ATP (3000 Ci/mM, NEN-Dupont) and 20 McKnight. A 539-bp fragment of the human b-actin was
units of T4 polynucleotide kinase (New England Biolabs, made by reverse-transcription polymerase chain reaction
Beverly, MA) in a 20 ml volume. The labeled oligonucle- using a sense primer 59-GTGGGGCGCCCCAGGCACCA
otide was mixed with twofold excess of cold antisense ol- (nt 144163) and an antisense primer 59-TCCTTAATGT-
igonucleotide, boiled at 1008C for 10 min, and slowly al- CACGCACGATTTC (nt 660683). All restriction diges-
lowed to cool to room temperature (RT). The double- tion fragments were purified from gel using the Jetsorb
stranded probe was then subjected to electrophoresis on a DNA purification kit (Genomed GmbH, Germany).
20% polyacrylamide gel in 13 TBE (0.09 M Tris, 0.09 M cDNA probes were made using 2550 ng of the insert,
boric acid, and 2 mM EDTA), and the wet gel was wrapped 50 mCi of a-32P-dCTP (3000 Ci/mM, NEN-Dupont), and 2
in plastic wrap and exposed for autoradiography. The band U of Klenow polymerase using the Multi-prime DNA-la-
was excised from the gel, and the labeled probe was ex- beling kit (Amersham Pharmacia Biotech, Piscataway, NJ).
tracted in a buffer containing 10 mM Tris-HCl, pH 8.0; 1 Unincorporated free 32P was removed using a spin column
mM EDTA; and 1 M NaCl at 378C overnight. After a phe- (Bio-Spin P30, Bio-Rad).
nol:chloroform extraction, the probe was precipitated with The membranes were washed once briefly in 23 SSC
100% ethanol in the presence of carrier tRNA (10 mg). The and were processed further for hybridization. Prehybridi-
labeled probe was recovered by centrifugation at 21 000 3 zation was carried out for 12 h in 6 ml of buffer containing
g for 15 min, washed once in 70% ethanol, dried, and dis- 50% de-ionized formamide, 53 Denhardt solution, 53 sa-
solved in minimal amount of sterile water. The radioactivity line-sodium phosphate-EDTA buffer, 1% SDS, 0.5 mg/ml
was measured using a Beckman scintillation counter. of denatured salmon sperm DNA, and 0.1 mg per ml of
The DNA-binding reaction was carried out in 30 ml vol- tRNA at 428C in a Hybaid oven with roller bottles (250 3
ume containing 5 mg of nuclear proteins from HepG2 cells 35 mm). Labeled probe in 4 ml of prehybridization buffer
or 30 to 40 mg of nuclear proteins from villous explants was added to the membrane (2 3 106 cpm per milliliter),
and 0.1 mg of denatured calf thymus DNA in 10 mM Tris and the hybridization was carried out as usual for 1224 h.
HCl, pH 7.5; 50 mM KCl; 50 mM NaCl; 1 mM MgCl2; 1 The membranes were washed in 23 SSC-0.1% SDS twice
mM EDTA; 5 mM DTT; and 5% glycerol. After a 5-min for 5 min at RT and then were washed twice in the same
preincubation, 5 3 104 cpm (0.5 to 1 ng) of the probe was solution at 378C. The washing was carried out for another
added, and the incubation was carried out for another 15 15 min at 508C with 0.1 3 SSC-0.1% SDS when necessary.
min at RT. The reaction mixture was loaded onto a 5% non- The membranes were exposed to Kodak-Bio-max-AR film
denaturing polyacrylamide gel and electrophoresed in 0.3 (Eastman Kodak).
3 TBE for 3 h at a constant 200 volts at 48C. The gels The membranes were stripped in 0.1% SDS at 608C for
were dried and exposed for autoradiography. In the case of 1530 min and reprobed for another gene of interest or for
supershift assays, 2 mg of the HIF antibodies or IgG con- the housekeeping gene, b-actin, to correct for interlane
trols were added to the mix after the initial 15 min, and the loading variations. b-actin was chosen as a housekeeping
incubation was further carried out for another 30 min before gene because it has been reported to be relatively unaf-
loading onto the gel. The gels were dried on Whatman 3 fected by hypoxia [37, 38]. Our results supported this con-
filter paper (Whatman International Co., Maidstone, UK) tentionb-actin mRNA (hypoxia)/b-actin mRNA (nor-
with a cellophane support (Bio-Rad) using a gel dryer moxia) was 0.88 6 0.10 for term placental villous explants
(Fisher Biotech, Pittsburgh, PA) and were exposed to Ko- (n 5 four placentas). The signal on the film was quantitated
dak-Bio-max-AR film (Eastman Kodak, Rochester, NY). by densitometry using the Videk Harmony program, ver-
sion 4.03 (Videk Corporation, Rochester, NY).
Northern Blot Analysis
Western Blot Analysis
Total RNA was isolated from HepG2 cells, placental tis-
sues, and villous explants using RNAwiz (Ambion, Austin, Total proteins from placental tissues or villous explants
TX). After a brief wash in ice-cold PBS, HepG2 cells were were extracted using 4 volumes of 13 Laemmli buffer (50
directly lysed in RNAwiz (1 ml per 75 cm2 flask). Placental mM Tris HCl, pH 6.8, 2% SDS, 10% glycerol) containing
tissues and villous explants were first pulverized using a 7 M urea, 5 mM DTT, 0.5 mM PMSF, and 1 mg/ml each
Bessman tissue pulverizer (Spectrum Medical Industries, of protease inhibitors, leupeptin, aprotinin, and pepstatin.
Laguna Niguel, CA) and then homogenized in 10 volumes Tissues were homogenized with a Tekmar Tissuemizer for
562 RAJAKUMAR AND CONRAD

about 1 min at setting 6. The crude homogenate was cen- tissues were fixed in 10% formalin for at least 24 h and
trifuged at 10 600 3 g at 48C, and the supernatant was then dehydrated and embedded in paraffin. Seven-micron
subjected to mild sonication (microprobe, setting 4 for 5 sections were cut and mounted on Fisher Superfrost/Plus
sec). The supernatant was then stored in aliquots at 2808C. glass slides. After removal of the paraffin, the sections were
Protein estimation was carried out using the Bio-Rad re- subjected to antigen retrieval by using Antigen Unmasking
agent, and 150 mg protein was used for the Western anal- Solution (Vector Laboratories Inc., Burlingame, CA) and a
ysis. Nuclear proteins from HepG2 cells (510 mg) or from microwave oven. After quenching of endogenous peroxi-
villous explants (3050 mg) were also used. All prepara- dases using 1.0% hydrogen peroxide in methanol for 3 min
tions were finally boiled for 5 min and briefly centrifuged. and blocking with 1.5% normal horse serum for 20 min,
The proteins were separated on a SDS containing 6% poly- the tissues were incubated with the monoclonal antibodies
acrylamide gel (Mini-Protean II cell system, Bio-Rad) at for 2 h at room temperature. Preliminary studies showed
100 V for one hour. Total protein samples were loaded in that the optimal concentration for the H1a67 HIF-1a anti-
40 ml volume containing the desired amount of protein and body was 10 mg/ml. The other antibodies were provided as
5% b-mercaptoethanol and 0.005% pyronin (Sigma Chem- cell culture supernatants at 10 mg/ml, and they were applied
ical Co., St. Louis, MO) and nuclear proteins in 2030 ml directly to the tissue sections. For negative controls, the
volume containing 2% SDS and 5% b-mercaptoethanol. IgG2b was substituted for the H1a67 HIF-1a at the same
The proteins were next transferred to PVDF membranes concentration. For the other antibodies, RPMI cell culture
(Immobilon; Millipore, Bedford, MA) using a semidry medium containing 10% FCS, 0.002% sodium azide, and
transfer system (Enprotech; Integrated Transfer Systems, 10 mg/ml of IgG1k was used. Using the Vectastain Elite
Natick, MA) at a constant current of 1.0 mA per cm2. De- ABC Kit and 3,39-diaminobenzidine (DAB) Peroxidase
tection of proteins was carried out after blocking the mem- Substrate Kit (both from Vector), immunoreactive HIF-1a
branes with a 1% solution of Blocking Reagent (Boehringer and -2a were detected. All of the villous placental tissues
Mannheim, Indianapolis, IN) for 4 h and incubating with depicted in Figure 6 were run in the same immunohisto-
the primary antibody. An anti-HIF-1a monoclonal antibody chemical procedure and developed with the DAB peroxi-
and a rabbit polyclonal anti-HIF-2a were obtained from dase substrate for identical times (11.5 min). Tissue sec-
Novus Biologicals (Littleton, CO; see Immunohistochem- tions were lightly counterstained with Hematoxylin Gill 2
istry for further details). For HIF-1a, the antibody was di- (Sigma). After dehydration in ethanol and xylene solutions,
luted 1:200 in TBS (Tris-buffered saline) buffer containing a coverslip was applied using Cytoseal XYL (Stephens Sci-
0.1% Tween-20, and for HIF-2a, the antibody was diluted entific, Riverdale, NJ).
1:400 in PBS containing 0.1% Tween-20. The correspond-
ing IgG was substituted for the primary antibody at the Data Analysis
same concentration to generate negative control blots. The
membranes were washed in either TBS-T or PBS-T buffer The data in all graphs are presented as mean or mean 6
three times for 10 min each and then were incubated with SEM. They were analyzed by one- or two-factor univariate
alkaline phosphatase-conjugated secondary antibody (1: analysis of variance (ANOVA). When appropriate, post hoc
2500 dilution for HIF-1 a and 1:5000 for HIF-2 a and b- comparisons between individual group means were made
actin) for 1 h. The membranes were washed three times in by Fishers protected least-significant difference tests. A P
either TBS-T or PBS-T for 10 min each. They were further value of less than 0.05 was considered to be significant
washed in the appropriate buffer without Tween-20 for 10 [41].
min and were allowed to equilibrate in alkaline phosphatase
buffer (100 mM Tris, pH 9.5; 150 mM NaCl) for 5 min. RESULTS
Chemiluminescent detection was carried out using the We first investigated the expression and ontogeny of the
CDP-Star substrate (Boehringer Mannheim) diluted 1:200 major hypoxia inducible transcription factors in normal hu-
in the alkaline phosphatase buffer for 5 min. Membranes man placentas of different gestational ages. By Northern
were exposed for different times to Kodak Bio-maxAR analysis, HIF-1a and -2a mRNA were present in normal
film. placentas at all stages of gestation with greater variability
The membranes were stripped with a buffer containing in expression during early pregnancy (Fig. 1A). Overall,
62.5 mM Tris HCl, pH 6.8; 2% SDS; and 100 mM b-ME however, the HIF-1a mRNA levels did not change signif-
at 508C for 1530 min and reprobed with another antibody icantly with advancing gestational age when normalized ei-
of interest or with a monoclonal anti-b-actin antibody ther for b-actin (Fig. 1B) or 18S RNA (data not shown;
(Clone AC-15, Sigma; 1:1000 dilution) to correct for load- values of both P 5 NS by ANOVA). In contrast, HIF-2a
ing variations. mRNA showed a significant increase during gestation,
whether normalized for b-actin (Fig. 1B, P 5 0.001) or
Immunohistochemistry 18S RNA (data not shown, P , 0.02). Although b-actin
and HIF-1a mRNA did not change significantly with ges-
Two anti-HIF-1a monoclonal antibodies were used. One, tational age when normalized for 18S RNA (P 5 NS by
a generous gift from Dr. Helen Turley, is clone Hif28b (a ANOVA), there was a trend for increased levels of both at
mouse IgG1k) directed against amino acids 329530 of the 58 gestational wks. These observations indicate that both
molecule [39]. The other designated clone H1a67 (a mouse HIF-1a and -2a mRNA are constitutively expressed in the
IgG2b) was purchased from Novus Biologicals and is di- human placenta.
rected against amino acids 432528. Dr. Turley also pro- A representative example of the expression and ontog-
vided a HIF-2a antibodyclone EP190b (a mouse IgG1k) eny of HIF-1a and -2a protein by Western analysis is de-
directed against amino acids 535631 of the molecule [40]. picted in Figure 1C. The graphical summary of the data
There was no cross-reactivity of the HIF-1a antibodies with from all placentas (some tested on duplicate blots and the
HIF-2a, and vice versa [39] (see Results). values averaged) is presented in Figure 1D. As a positive
Immediately after extraction from the uterus, placental control, total protein from a normal-term villous explant
HIF-1a, -2a, AND -1b IN THE HUMAN PLACENTA 563

FIG. 1. A) Representative Northern blot depicting the expression and ontogeny of HIF-1a and -2a mRNA in the human placenta. Ten micrograms of
total RNA were electrophoresed and Northern analysis performed (see Materials and Methods). The 4.4-kilobase (kb) HIF-1a (top panel), 5.8-kb HIF-
2a (middle panel), and the 1.8 kb b-actin (bottom panel) mRNA that was used to normalize for loading variability are shown. Each lane contains total
RNA from a different placenta. B) Graphical summary of all data is presented in A (mean 6 SEM). The n indicates the number of different placentas
analyzed for each gestational age. There was a significant effect of gestational age on HIF-2a mRNA expression (P 5 0.001 by ANOVA). * P , 0.05
compared with all other gestational ages. Note that comparison of the abundance of mRNA for HIF-1a and -2a cannot be made because the specific
activity of the probes is unknown. C) Representative Western blot depicting the expression and ontogeny of HIF-1a and -2a protein in the human
placenta. Total protein was extracted in 7 M urea containing Laemmli buffer, electrophoresed on SDS containing 6% polyacrylamide gels, and subjected
to Western analysis as described in Materials and Methods. The 120-kDa HIF-1a, 115-kDa HIF-2a, and 43-kDa b-actin proteins are shown. The total
protein from a normal-term villous explant subjected to 46 h of hypoxia represents a positive control. The exposure times for HIF-1a and -2a,
respectively, were 30 and 3 min. Each lane contains total protein from a different placenta. D) Graphical depiction of all of the data for expression of
HIF-1a and -2a protein in the human placenta (mean 6 SEM). The n refers to the number of different placentas tested at each gestational age. There
was a significant effect of gestational age on both HIF-1a and -2a protein expression (P , 0.01 and , 0.005 by ANOVA, respectively). * P , 0.05
compared with 58 wk; 1 P , 0.05 compared with all other gestational ages. Note that comparison of the abundance of protein for HIF-1a and -2a
cannot be made because the relative efficacy of the antibodies is unknown.

exposed to 2% hypoxia in culture for 46 h was also using term villous explants (Fig. 3, A and B), that is, there
probed for HIF-1a and -2a (see below). A significant effect was no influence of hypoxia on either HIF-1a or -2a
of gestational age was observed for both HIF-1a and HIF- mRNA expression (P 5 NS by ANOVA). The HepG2 cells
2a (P , 0.01 and , 0.005 by ANOVA, respectively). The also failed to show any difference in the mRNA expression
protein expression was significantly greater at the earlier between normoxic and hypoxic conditions (Fig. 3A). Thus,
gestational stages. placental HIF expression does not appear to be regulated
To determine whether hypoxia regulates HIF expression by hypoxia at the level of mRNA.
in the human placenta and, if so, at the level of mRNA, In contrast, the expression of both HIF-1a and -2a pro-
protein, or both, we subjected villous explants to standard tein is regulated by hypoxia in the human placenta. Figure
tissue culture conditions (normoxia) or to an hypoxic en- 4, A and B, depicts the Western analyses using total and
vironment (1 or 2% O2; see Materials and Methods). As nuclear proteins extracted from villous explants, respec-
shown in Figure 2, A and B, there was no consistent effect tively (see Materials and Methods). Proteins of 120 and
of hypoxia on either HIF-1a or -2a mRNA expression in 115 kDa representing HIF-1a and -2a, respectively, were
villous explants derived from first-trimester placentas of 5, enhanced by 2% hypoxia. Term villous explants that were
7, or 12 gestational wk. Additional placental villous ex- exposed to 1% hypoxia showed even further increases in
plants from first-trimester placentas were tested. After 6 h HIF-1a but not in HIF-2a expression, which was appar-
of incubation in normoxic or hypoxic conditions, normal- ently already maximal at 2% O2 (Fig. 4A). By reprobing
ized HIF-1a mRNA was 2.29 and 2.07, 2.18 and 2.10, and the same membrane with an antibody directed against b-
1.97 and 1.63, respectively, for the villous explants from actin, uniform loading was documented. Nuclear extracts
7-, 9-, and 11-wk placentas. Normalized HIF-2a in the from HepG2 cells were used as a positive control (Fig. 4A).
same tissues was 1.51 and 1.68, 1.55 and 1.83, and 1.50 To establish the DNA-binding activity of HIF, electro-
and 1.39, respectively. Comparable results were obtained phoretic mobility shift assays (EMSA) were performed. A
564 RAJAKUMAR AND CONRAD

FIG. 2. A) Expression of HIF-1a and -2a mRNA in first-trimester villous FIG. 3. A) Expression of HIF-1a, -2a, and -1b mRNA in term villous
explants subjected to normoxia (N) or hypoxia (H; 2% O2). Ten mg of total explants subjected to normoxia (N) or hypoxia (H). Twenty micrograms
RNA were electrophoresed, and Northern analysis was performed (see of total RNA was electrophoresed and analyzed by Northern analysis (see
Materials and Methods). The 4.4-kb HIF-1a (top panel), 5.8-kb HIF-2a Materials and Methods). The 4.4-kb HIF-1a (top panel), 5.8-kb HIF-2a
(middle panel), and 1.8-kb b-actin (bottom panel) mRNA are shown. B) (second panel), 2.6-kb HIF-1b (third panel), and 1.8-kb b-actin (bottom
Graphical illustration of the data presented in A (mean values). Note the panel) mRNA are shown. This blot is representative of identical experi-
inclusion of data from an additional first-trimester (7-wk) placenta. There ments conducted on four placentas. Northern analysis of total RNA from
was no effect of hypoxia on mRNA expression. HepG2 cells exposed to normoxic and hypoxic conditions for 4 h is also
shown. B) Graphical depiction of the data shown in A (mean 6 SEM).
There was no significant effect of hypoxia on HIF mRNA expression by
ANOVA.
distinct HIF complex that moves above a constitutively ex-
pressed complex was observed in the positive control
HepG2 cells and also in the placental villous explants ex- antibodies to HIF-1a and -1b in supershift assays (Fig. 5B).
posed to hypoxia (Fig. 5, AD). The signal in the placental Substitution of the appropriate IgG control for the HIF-1a
villous explants was generally less than the HepG2 cells, and -1b antibodies at the same concentration did not affect
even though we used considerably more villous nuclear ex- the mobility of the HIF complex.
tract in each reaction mixture3040 compared with 5 The amount of HIF DNA-binding activity varied with
10 mg. We considered that this discrepancy might be due the degree of hypoxia (see Fig. 5C). That is, term villous
to an abundance of CREB/ATF that may compete with explants exposed to 1% O2 demonstrated more HIF DNA-
HIF-1a for the labeled oligonucleotide. Therefore, we add- binding activity than did those incubated with 2% O2. Co-
ed cold AP1/CREB consensus oligonucleotide as a com- balt chloride duplicated the hypoxia response in villous ex-
petitor. This maneuver somewhat enhanced the DNA-bind- plants maintained under normoxic incubation conditions
ing activity of HIF-1a and decreased the binding activity (Fig. 5C). The effect of preincubating HepG2 cells and term
of the constitutively expressed band (putatively CREB/ villous explants with actinomycin D or cyclohexamide is
ATF). The HIF complex for the HepG2 cells was frequently shown in Figure 5D. Although actinomycin D had little or
a doublet, whereas the HIF complex observed for the nu- no effect on HIF DNA-binding activity, cyclohexamide
clear extracts from villous explants generally migrated with completely abolished the binding. Finally, as a negative
the faster-moving lower band of the HepG2 complex. Spec- control, SP1 consensus oligonucleotide was employed in
ificity of the HIF DNA binding was established by utilizing the mobility-shift assay to further determine whether the
HIF-1a, -2a, AND -1b IN THE HUMAN PLACENTA 565

FIG. 4. A) Expression of HIF-1a and -2a protein in villous explants ex-


posed to normoxia (N) or hypoxia (H). Total protein (150 mg) that was
extracted in 7 M urea containing Laemmli buffer was separated on SDS
containing 6% polyacrylamide gels and subjected to Western analysis
(see Materials and Methods). A representative blot of three experiments
is shown with the 120-kDa HIF-1a, 115-kDa HIF-2a, and 43-kDa b-actin
protein. The last two lanes contain nuclear protein from HepG2 cells (10 FIG. 5. A) Electrophoretic mobility shift assays representative of identical
mg per lane) that serves as a positive control. H2%, 2% O2; H1%, 1% experiments conducted on four placentas employing nuclear extracts
O2; otherwise, H alone denotes 2% O2. B) Nuclear proteins were extract- from term villous explants and HepG2 cells (positive control) exposed to
ed from HepG2 cells (10 mg per lane) and villous explants (50 mg per normoxia (N) or hypoxia (H). The identical trends were seen in all ex-
lane), as explained in Materials and Methods and subjected to Western periments. A double-stranded oligonucleotide containing the hypoxia re-
analysis. Nuclear proteins from 7- and 12-wk and term villous explants sponse element from the erythropoietin gene was end-labeled with 32P-g
that were exposed to normoxia (N) and hypoxia (H) for 6 h were analyzed ATP. Approximately 50 000 cpm was mixed with 510 mg and 3040 mg,
along with the nuclear proteins from the HepG2 cells, the latter serving respectively, of nuclear proteins from the HepG2 cells and term villous
as a positive control. explants. The protein-DNA complex was separated on a 5% nondenatur-
ing gel in 0.33 TBE. The gels were dried and exposed for autoradiography.
Because the HIF complex observed in villous explant tissue was weaker,
hypoxia response was specific. The ubiquitous transcription 110 ng of a mixture of double-stranded AP1 and CREB was added to
factors SP1 and SP3 present in nuclear extracts of HepG2 the binding reaction, which moderately enhanced the intensity of the HIF
cells and term villous explants were not affected by hypoxia complex by reducing the CREB/ATF complex. All experiments were there-
fore carried out in the presence of these oligonucleotides (see Materials
(Fig. 5E). and Methods for additional details). NS, Nonspecific band; C, constitu-
Cells that are exposed to hypoxia turn on a battery of tively expressed band; HIF, mobility shift caused by HIF-1a and HIF-1b
genes as an adaptive response in many instances via the heterodimer. B) Supershift assays using HIF-specific antibodies. Electro-
HIF pathway, for instance, glucose transporter-1 (Glut-1) phoretic mobility shift assays were performed using nuclear extracts from
[11]. Therefore, we measured Glut-1 mRNA. The same HepG2 cells (positive control) and from 7-wk and term villous explants.
membrane depicted in the Northern blot in Figure 3A was After an initial 15-min incubation, 2 mg of HIF-1a or -1b (data not shown)
antibody was added to the reaction mixture, and the incubation was con-
stripped and reprobed for Glut-1 expression. Although the tinued for another 3060 min on ice. Mouse IgG2b and rabbit IgG were
HepG2 cells exposed to hypoxia showed some increase in substituted for the HIF-1a and -1b antibodies, respectively, as negative
Glut-1 mRNA, the villous explant response was more controls. The complexes were separated on gel and analyzed by autora-
marked, and at each time point, Glut-1 mRNA was clearly diography (see Materials and Methods for further details). C) Electropho-
more abundant in the hypoxic tissues (Fig. 5F). retic mobility shift assays of nuclear extracts prepared from term villous
Because placental tissue is comprised of several cell explants exposed for 6 h to normoxia (N), hypoxia at 2% O2 (H2%),
hypoxia at 1% O2 (H1%), or CoCl2. D) Electrophoretic mobility shift as-
types, we employed immunohistochemistry to discern lo- says of nuclear extracts prepared from HepG2 cells and term villous ex-
calization of HIF-1a and -2a. Additionally, this technique plants treated with actinomycin D (ACT) or cyclohexamide (CHX) prior
provides information regarding cytoplasmic and/or nuclear to and during subsequent incubation in normoxic (N) or hypoxic (H) con-
expression. All results presented in Figure 6 are represen- ditions (see Materials and Methods for additional details). E) Electropho-
tative of at least three placentas studied during the first retic mobility shift assays showing the presence of the ubiquitous tran-
trimester and term. Figure 6, af, depict the results for HIF- scription factors SP1 and SP3 in the nuclear extracts of HepG2 cells and
term villous explants. The levels of these transcription factors were not
1a in first trimester placentas. This transcription factor was regulated by hypoxia. F) Northern analysis depicting the expression of the
observed in the syncytiotrophoblast, villous cytotropho- 2.8-kb glucose transporter-1 mRNA in HepG2 cells and term villous ex-
blast, fetoplacental vascular endothelium, and possibly oth- plants exposed to normoxic (N) and hypoxic (H) incubation conditions.
er cell types in the villous core. Comparable results were The blot from Figure 3A was stripped and reprobed for the Glut-1 ex-
observed for term villous placenta (Fig. 6, gi), except that pression shown herefor b-actin, see Figure 3A.
at this gestational stage, the villous cytotrophoblast is rare
566 RAJAKUMAR AND CONRAD

FIG. 6. Localization of immunoreactive HIF-1a and HIF-2a in the first-trimester and term villous placenta (brown staining). ac) Serial sections of a
7-wk placenta for immunolocalization of HIF-1a using clones H1a67 monoclonal IgG2b and Hif28b monoclonal IgG1k antibodies in a and c, respectively.
Substitution of the primary antibody with mouse IgG2b or IgG1k as a negative control, respectively, is shown in the upper and lower panels of b. df)
Serial sections of a 9.5-wk placenta for immunolocalization of HIF-1a again using the two clones of HIF-1a antibodies in (d) and (f), respectively. The
negative controls using IgG2b and IgG1k are depicted in the upper and lower panels of e. gi) Serial sections of a term placenta for immunolocalization
of HIF-1a, again using the clones H1a67 and Hif28b monoclonal antibodies in g and i, respectively. The negative controls using IgG2b and IgG1k are
shown in the upper and lower panels of h, respectively. jl) Tissue sections of a 9.5-wk (j) and normal-term placenta (l) for immunolocalization of HIF-
2a using the clone EP190b monoclonal IgG1k antibody. Substitution of the primary antibody with mouse IgG1k is shown in the upper and lower panels
of k for the 9.5-wk and term placenta, respectively. The syncytiotrophoblast is denoted by the thin arrow, the villous cytotrophoblast by the arrow head,
and fetoplacental blood vessels by the thick arrow. Original magnification: 3200.
HIF-1a, -2a, AND -1b IN THE HUMAN PLACENTA 567

and not easily identified. Significantly, the data were similar the human placenta by oxygen, we investigated cultured
for the two different HIF-1a monoclonal antibodies em- villous explants subjected to a 20%, 2%, or 1% oxygen
ployed, although the immunostaining using clone H1a67 atmosphere as previously described [31]. A major obser-
was generally more intense (see figure legend and Materi- vation was that HIF-1a, -1b, and -2a were not regulated
als and Methods for further details). Figure 6, jl, portrays by oxygen at the level of mRNA in either first trimester
the findings for HIF-2a in the first trimester (Fig. 6j and (Fig. 2, A and B) or term (Fig. 3, A and B) villous explants,
the top of Fig. 6k), and term placentas (the bottom of Fig. as assessed by Northern analysis. Nor was regulation of the
6k and Fig. 6l). The localization of HIF-2a was generally HIF message by hypoxia observed in the HepG2 cells (Fig.
overlapping with HIF-1a. Both nuclear and cytoplasmic lo- 3A). Although there may have been some diminution of the
calization are evident for the two transcription factors. hypoxia-induced HIF-1a DNA-binding activity by actino-
mycin D for the HepG2 cells as determined by EMSA, no
DISCUSSION such decrease was observed for the term villous explants
Despite the close link between placental hypoxia and (Fig. 5D). Thus, the studies with actinomycin D and HIF-
normal placental development and various placental pa- 1a DNA binding activity are consistent with the Northern
thologies, relatively little is known about the molecular analysis, at least for placental villous explants. Although
mechanisms of oxygen transduction by this organ. Accord- some investigators have reported hypoxia modulation of
ingly, our objectives were to investigate the ontogeny of HIF mRNA, others have not, and the phenomenon may
expression of the hypoxia-inducible transcription factors in depend on the tissue or cells being investigated, on incu-
the human placenta, as well as their regulation by oxygen bation conditions, and on the degree of hypoxia ([44], and
using villous explant cultures. see citations in [11], as well as present results).
A major finding was that HIF-2a mRNA increased sig- In contrast, of major significance was that both HIF-1a
nificantly with gestational age, whereas HIF-1a remained and -2a proteins were potently induced by hypoxia in first
unchanged as assessed by Northern analysis (Fig. 1, A and trimester and term villous explants, as well as in HepG2
B). An increase in HIF-2a mRNA per cell, an increase in cells (Fig. 4, A and B). Presumably, this event was sec-
the number of villous cells expressing the message, or both ondary to protein stabilization [14, 1618] and increased
could account for the phenomenon. Because HIF-2 a translation (e.g., [11, 24]). Our experiments with cycloh-
mRNA may be expressed preferentially in endothelial cells examide and HIF-1a DNA-binding activity were supportive
[13, 14], the development and maturation of the fetopla- of the latter mechanism (Fig. 5D, see below), but because
cental vasculature in the villous core with advancing ges- the translation of all proteins was inhibited by the drug, this
tational age [4] might explain the finding. Ultimately, in methodological approach was nonspecific and did not ex-
situ hybridization is needed to resolve this issue. clude an important contribution of protein stabilization. In
Another major finding was that the abundance of both addition, more HIF-1a protein was expressed in response
HIF-1a and -2a protein in the human placenta significantly to 1.0% rather than 2.0% hypoxia in term villous explants.
decreased with gestational age as determined by Western The HIF-1a DNA binding activity on EMSA was also more
analysis (Fig. 1, C and D). In fact, there was an inverse intense in term villous explants subjected to the 1.0% hyp-
relationship between HIF-2a mRNA and protein expres- oxic incubation (Fig. 5C). This inverse relationship between
sion, with the former significantly rising and the latter de- the degree of hypoxia and the production of HIF-1a protein
clining with gestational age (compare Fig. 1, B and D). and DNA-binding activity, at least down to 0.5% O2, has
Because HIF-1a and -2a protein, but not mRNA, are in- been previously described [45]. In contrast, no further in-
duced in the placenta by hypoxia (see below), the finding crease was observed for the HIF-2a protein, which is also
of increased protein expression in the first trimester is con- consistent with previous work suggesting that HIF-2a may
sistent with the low intervillous blood flow and physiolog- be more active than HIF-1a under less severely hypoxic
ical hypoxia that has been reported during the early stages conditions [39].
of placental development [2, 3]. Interestingly, despite the Using EMSA, we documented the corresponding DNA
establishment of intervillous blood flow after the first tri- binding activity of HIF-1a for first-trimester and term vil-
mester, both HIF-1a and -2a proteins were still detected in lous explants, as well as for the HepG2 cells (Fig. 5). A
second and third trimester placentas. On the one hand, per- constitutive band was also present that may represent
sistent protein expression may be partly a consequence of CREB/ATF DNA-binding activity [46]. The authenticity of
the relatively low PO2 measured in the intervillous space the HIF-1a DNA-binding activity was verified by super-
even after the establishment of intervillous blood flow60 shifts with antibodies specific for HIF-1a and -1b (Fig. 5B).
torr (vs. arterial PO2 of 75100 torr)which was compa- The HIF complex was a doublet for the HepG2 cells,
rable to the endometrial PO2 [3]. On the other hand, various whereas the DNA-binding activity of the villous explants
growth factors including IGF-I and EGF [21, 22] may con- was a single band that aligned with the lower, faster mo-
tribute to this paradoxical persistence of HIF-1a and -2a bility band of the doublet observed for the HepG2 cells
protein expression after the first trimester and possibly to (best portrayed in Fig. 5D). Maxwell et al. showed that the
the elevated levels observed during the first trimester. In- upper, slower-mobility band of the HIF/DNA complex was
deed, the expression of IGF-I and -II, as well as of the IGF- supershifted by a von Hippel-Lindau (VHL) protein anti-
binding proteins by the placenta has been described [42]. body [47]. Thus, an interaction between VHL protein and
Our findings are perhaps reminiscent of those recently re- the HIF/DNA complex may not occur in the human pla-
ported by Iyer and colleagues that indicate that mouse em- centa during hypoxic conditions comparable to the treat-
bryonic stem cells showed relatively high levels of HIF-1a ment with cobalt chloride ([47] and Fig. 5C). Further in-
expression under nonhypoxic conditions [25]. Furthermore, vestigation is needed to substantiate this possibility.
the presence of HIF-1a and -2a protein in the human pla- We were unable to identify HIF-2a-binding activity on
centa may explain the constitutive expression of erythro- EMSA despite the presence of mRNA and protein in the
poietin by this organ [43]. villous explants, the latter of which was increased by hyp-
In order to evaluate the regulation of HIF-1a and -2a in oxia as described above. Indeed, the HIF complex that we
568 RAJAKUMAR AND CONRAD

observed was consistently supershifted by the HIF-1a an- Regulation. Oxford, UK: BIOS Scientific Publishers, Ltd.; 1999: 25
tibody, and there were no other candidate bands. Clearly, 45.
12. Wang GL, Jiang B-H, Rue EA, Semenza GL. Hypoxia-inducible fac-
supershift studies using the HIF-2a antibodies are needed. tor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular
To our knowledge, there are no published reports identi- O2 tension. Proc Natl Acad Sci U S A 1995; 92:55105514.
fying endogenous HIF-2a DNA-binding activity by EMSA. 13. Tian H, McKnight SL, Russell DW. Endothelial PAS domain protein
It is possible that the binding conditions are different, and 1 (EPAS1), a transcription factor selectively expressed in endothelial
further work is needed to clarify this important issue. cells. Genes Dev 1997; 11:7282.
Because placental villi are composed of many cell types, 14. Ema M, Taya S, Yokotani N, Sogawa K, Matsuda Y, Fjuii-Kuriyama
Y. A novel bHLH-PAS factor with close sequence similarity to hyp-
we used immunohistochemistry to determine cellular lo- oxia-inducible factor 1a regulates the VEGF expression and is poten-
calization of HIF-1a and -2a (Fig. 6). The technique also tially involved in lung and vascular development. Proc Natl Acad Sci
provided insight into nuclear and/or cytoplasmic localiza- U S A 1997; 94:42734278.
tion. The major findings for the first trimester and term 15. Zhu H, Bunn HF. Oxygen sensing and signaling: impact on the reg-
placentas were 1) the transcription factors showed overlap- ulation of physiologically important genes. Respir Physiol 1999; 115:
ping expression in the syncytiotrophoblast, villous cytotro- 239247.
16. Huang LE, Gu J, Schau M, Bunn HF. Regulation of hypoxia-inducible
phoblast, fetoplacental vasculature, and possibly in other factor 1a is mediated by an O2-dependent degradation domain via the
villous core cells and 2) they demonstrated both nuclear ubiquitin-proteasome pathway. Proc Natl Acad Sci U S A 1998; 95:
and cytoplasmic localization. The conclusions on HIF-1 a 79877992.
may be particularly reliable because comparable results 17. Salceda S, Caro J. Hypoxia-inducible factor 1a (HIF-1a) protein is
were obtained with two different HIF-1a monoclonal an- rapidly degraded by the ubiquitin-proteasome system under normoxic
tibodies. conditions. J Biol Chem 1997; 272:2264222647.
18. Huang LE, Arany Z, Livingston DM, Bunn HF. Activation of hypoxia-
The induction of glucose transporter-1 by hypoxia has inducible transcription factor depends primarily upon redox-sensitive
been previously shown to be mediated by HIF-1 [11]. In- stabilization of its a subunit. J Biol Chem 1996; 271:3225332259.
deed, the activation of HIF by hypoxia in the villous ex- 19. Ema M, Hirota K, Mimura J, Abe H, Yodoi J, Sogawa K, Poellinger
plants was associated with the induction of Glut-1 mRNA L, Fujii-Kuriyama Y. Molecular mechanisms of transcription activa-
(Fig. 5F). Clearly, further investigation is required to estab- tion by HLF and HIF1a in response to hypoxia: their stabilization and
lish additional links between the hypoxia-inducible tran- redox signal-induced interaction with CBP/p300. EMBO J 1999; 18:
19051914.
scription factors and normal placental development, as well 20. Arany Z, Huang LE, Eckner R, Bhattacharya S, Jiang C, Goldberg
as placental pathologies. MA, Bunn HF, Livingston DM. An essential role for p300/CBP in the
cellular response to hypoxia. Proc Natl Acad Sci U S A 1996; 93:
ACKNOWLEDGMENTS 1296912973.
21. Zelzer E, Levy Y, Kahana C, Shilo B-Z, Rubinstein M, Cohen B.
Insulin induces transcription of target genes through the hypoxia-in-
We thank Dr. Helen Turley and her colleagues for their magnanimous ducible factor HIF-1a/ARNT. EMBO J 1998; 17:50855094.
contribution of HIF-1a and -2a antibodies, and we also thank Dr. Gregg 22. Feldser D, Agani F, Iyer NV, Pak B, Ferreira G, Semenza GL. Recip-
Semenza and Dr. Steven McKnight for their generous gifts of the HIF-1a rocal positive regulation of hypoxia-inducible factor 1a and insulin-
and HIF-2a cDNA probes, respectively. The authors also thank Rob Smith like growth factor 2. Cancer Res 1999; 59:39153918.
for his assistance in the procurement of placentas and Sue Davis for out- 23. Jiang B-H, Agani F, Passaniti A, Semenza GL. V-SRC induces ex-
standing clerical support. pression of hypoxia-inducible factor 1 (HIF-1) and transcription of
genes encoding vascular endothelial growth factor and enolase 1: in-
REFERENCES volvement of HIF-1 in tumor progression. Cancer Res 1997; 57:5328
5335.
1. Khong TY, Robertson WB. Spiral artery disease. In: Coulam CG, 24. Wang GL, Semenza GL. Desferrioxamine induces erythropoietin gene
Faulk WP, McIntyre JA (eds.), Immunological Obstetrics. New York: expression and hypoxia-inducible factor 1 DNA-binding activity: im-
W.W. Norton & Company; 1992: 492501. plications for models of hypoxia signal transduction. Blood 1993; 82:
2. Carter AM. When is the maternal placental circulation established in 36103615.
man? Placenta 1997; 18:8387. 25. Iyer NV, Kotch LE, Agani F, Leung SW, Laughner E, Wenger RH,
3. Rodesch F, Simon P, Donner C, Jauniaux E. Oxygen measurements in Gassmann M, Gearhart JD, Lawler AM, Yu AY, Semenza GL. Cel-
endometrial and trophoblastic tissues during early pregnancy. Obstet lular and developmental control of O2 homeostasis by hypoxia-induc-
Gynecol 1992; 80:283285. ible factor 1a. Genes Dev 1998; 12:149162.
4. Benirschke K, Kaufmann P. Pathology of the Human Placenta, 3rd 26. Tian H, Hammer RE, Matsumoto AM, Russell DW, McKnight SL.
ed. New York: Springer-Verlag; 1995. The hypoxia-responsive transcription factor EPAS1 is essential for
5. Genbacev O, Joslin R, Damsky CH, Polliotti BM, Fisher SJ. Hypoxia catecholamine homeostasis and protection against heart failure during
alters early gestation human cytotrophoblast differentiation/invasion embryonic development. Genes Dev 1998; 12:33203324.
in vitro and models the placental defects that occur in preeclampsia. 27. Maltepe E, Schmidt JV, Baunoch D, Bradfield CA, Simon MC. Ab-
J Clin Invest 1996; 97:540550. normal angiogenesis and responses to glucose and oxygen deprivation
6. Genbacev O, Zhou Y, Ludlow JW, Fisher SJ. Regulation of human in mice lacking the protein ARNT. Nature 1997; 386:403407.
placental development by oxygen tension. Science 1997; 277:1669 28. Wiener CM, Booth G, Semenza GL. In vivo expression of mRNAs
1672. encoding hypoxia-inducible factor 1. Biochem Biophys Res Commun
7. Lindheimer MD, Roberts JM, Cunningham FG. Chesleys Hyperten- 1996; 225:485488.
sive Disorders in Pregnancy, 2nd ed. Stamford, CT: Appleton & 29. Seligman SP, Nishiwaki T, Kadner SS, Dancis J, Finlay TH. Hypoxia
Lange; 1999. stimulates ecNOS mRNA expression by differentiated human tropho-
8. Conrad KP, Benyo DF. Placental cytokines and the pathogenesis of blasts. Ann NY Acad Sci 1997; 828:180187.
preeclampsia. Am J Reprod Immunol 1997; 37:240249. 30. Janatpour MJ, Utset MF, Cross JC, Rossant J, Dong J, Israel MA,
9. Lim K-H, Zhou Y, Janatpour M, McMaster M, Bass K, Chun S-H, Fisher SJ. A repertoire of differentially expressed transcription factors
Fisher SJ. Human cytotrophoblast differentiation/invasion in abnormal that offers insight into mechanisms of human cytotrophoblast differ-
pre-eclampsia. Am J Pathol 1997; 151:18091818. entiation. Dev Genet 1999; 25:146157.
10. Caniggia I, Grisaru-Gravnosky S, Kuliszewsky M, Post M, Lye SJ. 31. Benyo DF, Miles TM, Conrad KP. Hypoxia stimulates cytokine pro-
Inhibition of TGF-b3 restores the invasive capability of extravillous duction by villous explant from the human placenta. J Clin Endocrinol
trophoblasts in preeclamptic pregnancies. J Clin Invest 1999; 103: Metab 1997; 82:15821588.
16411650. 32. Wolff M, Fandrey J, Jelkmann W. Microelectrode measurements of
11. Wenger RH, Gassmann M. HIF-1 and the molecular response to hyp- pericellular PO2 in erythropoietin-producing human hepatoma cell
oxia in mammals. In: Storey KB (ed.), Environmental Stress and Gene cultures. Am J Physiol 1993; 265:C1266-C1270.
HIF-1a, -2a, AND -1b IN THE HUMAN PLACENTA 569

33. Metzen E, Wolff M, Fandrey J, Jelkmann W. Pericellular PO2 and O2 Harris AL. The distribution of the hypoxia inducible factors HIF-1a
consumption in monolayer cell cultures. Respir Physiol 1995; 100: and HIF-2a in normal and pathological tissues. Am J Pathol 2000;
101106. (in press).
34. Gilson GJ, Mosher MD, Conrad KP. Systemic hemodynamics and 41. Zar JH. Biostatistical Analysis, 2nd ed. Englewood Cliffs, NJ: Pren-
oxygen transport during pregnancy in chronically-instrumented, con- tice Hall; 1984.
scious rats. Am J Physiol 1992; 263:H1911-H1918. 42. Han VKM, Bassett N, Walton J, Challis JRG. The expression of in-
35. Maxwell PH, Pugh CW, Ratcliffe PJ. Inducible operation of the eryth- sulin-like growth factor (IGF) and IGF-binding protein (IGFBP) genes
ropoietin 39 enhancer in multiple cell lines: evidence for a widespread in the human placenta and membranes: evidence for IGF-IGFBP in-
oxygen-sensing mechanism. Proc Natl Acad Sci U S A 1993; 90: teractions at the feto-maternal interface. J Clin Endocrinol Metab
24232427. 1996; 81:26802693.
36. Semenza GL, Wang GL. A nuclear factor induced by hypoxia via de 43. Conrad KP, Benyo DF, Westerhausen-Larson A, Miles TM. Expression
novo protein synthesis binds to the human erythropoietin gene en- of erythropoietin by the human placenta. FASEB J 1996;10:760766.
hancer at a site required for transcriptional activation. Mol Cell Biol 44. Yu AY, Frid MG, Shimoda LA, Wiener CM, Stenmark K, Semenza
1992; 12:54475454. GL. Temporal, spatial, and oxygen-regulated expression of hypoxia-
37. Graven KK, Troxler RF, Kornfeld H, Panchenko MV, Farber HW. inducible factor-1 in the lung. Am J Physiol 1998; 275:L818L826.
Regulation of endothelial cell glyceraldehyde-3-phosphate dehydro- 45. Jiang B-H, Semenza GL, Bauer C, Marti HH. Hypoxia-inducible fac-
genase expression by hypoxia. J Biol Chem 1994; 269:2444624453. tor 1 levels vary exponentially over a physiologically relevant range
38. Zhong H, Simons JW. Direct comparison of GAPDH, b-actin, cyclo- of O2 tension. Am J Physiol 1996; 271:C1172C1180.
philin, and 28S rRNA as internal standard for quantifying RNA levels 46. Kvietikova I, Wenger RH, Marti HH, Gassmann M. The transcription
under hypoxia. Biochem Biophys Res Commun 1999; 259:523526. factors ATF-1 and CREB-1 bind constituitively to the hypoxia-induc-
39. Wiesener MS, Turley H, Allen WE, William C, Eckardt K-U, Talks ible factor-1 (HIF-1) DNA recognition site. Nucleic Acids Res 1995;
KL, Wood SM, Gatter KC, Harris AL, Pugh CW, Ratcliffe PH, Max- 23:45424550.
well PH. Induction of endothelial PAS domain protein-1 by hypoxia: 47. Maxwell PH, Wiesener MS, Chang G-W, Clifford SC, Vaux EC,
characterization and comparison with hypoxia-inducible factor-1a. Cockman ME, Wykoff CC, Pugh CW, Maher ER, Radcliffe PJ. The
Blood 1998; 92:22602268. tumour suppressor protein VHL targets hypoxia-inducible factors for
40. Talks KL, Turley H, Gatter KC, Maxwell PH, Pugh CW, Radcliffe PJ, oxygen-dependent proteolysis. Nature 1999; 399:271275.

You might also like