You are on page 1of 17

Molecular and Cellular Endocrinology 228 (2004) 103–119

Review
Endocrine cell lines from the placenta
M.H.F. Sullivan∗
Department of Obstetrics and Gynaecology, Faculty of Medicine, Wolfson and Weston Research Centre for Family Health, Institute of Reproductive and
Developmental Biology, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK

Received 16 January 2003; accepted 31 March 2003

Abstract

Cell-lines derived from human placenta and chorion have been used extensively to model the endocrine functions of human trophoblast. In
general terms, the endocrine functions of the primary cells and tissues are at least partially replicated within the cell-lines, suggesting that they
may be used as appropriate models. There are, however, two major provisos that compromise this generalisation. Firstly, the endocrine function
of placenta represents a complex interaction between cytotrophoblast, syncytiotrophoblast and multiple regulators, so a single cell population
digested from the normal environment is unlikely to represent this. Secondly, the characterisation of primary trophoblast populations and of
cell-lines is incomplete, complicating the assignment of functions to trophoblast populations. Despite these difficulties, useful information
has been obtained from the available cell-lines, regardless of whether they have arisen spontaneously, been transformed in vitro, or derived
from cancers in vivo.
© 2004 Elsevier Ireland Ltd. All rights reserved.

Keywords: Endocrine cell lines; Placenta; Pregnancy

1. Defining the endocrinology of normal human non-pregnant adult. On this basis factors that have a regula-
placenta tory role in placental endocrine function, but are not present
at increased levels in maternal blood during pregnancy may
The placenta has a very wide range of functions in preg- be included in this review. For a fuller treatment of the HPA
nancy, such that it has been described as an experimental and HPG axes, readers should refer to the appropriate chap-
system in its own right, independently of its interactions ters elsewhere in this special issue of Molecular and Cellu-
with fetal and maternal physiology. In seeking to identify lar Endocrinology, as well as general reviews.
the endocrine functions of the human placenta, it is nec- The human placenta does not generally seem to be de-
essary to consider what might be an appropriate defini- pendent on these maternal endocrine systems, although it
tion. Classically, endocrinology has been defined as “The can regulate the maternal HPA and HPG axes, and there
study of the system of glands and tissues that produce and is cross-talk between the placental and maternal systems
secrete hormones”—the latter being active regulatory sub- (Glinoer et al., 1990; Lockwood et al., 1996; Petraglia et al.,
stances formed in one part of the body and carried by the 1996, 1998; Miller, 1998). It has the capacity to produce
blood to another, where they signal the coordination of cel- factors that typify hypothalamic, pituitary and ovarian func-
lular functions. Placental function does not necessarily fit tions, and the primary placental sources are listed in Table 1;
within these definitions, as placental products may have in- it must be pointed out that some of these factors are in-
trauterine targets as well as extrauterine targets, and only cluded as regulators of placental function, and hence of the
the latter would necessarily include the blood-borne aspect. outcome of pregnancy, rather than being part of the HPA or
In this review I propose to include all factors that are con- HPG axes. These will be considered only in so far as they
sidered to part of the hypothalamic-pituitary-adrenal (HPA) contribute to the regulation of endocrine placental functions.
axis or the hypothalamic-pituitary-gonadal (HPG) axis in the A series of papers and reviews have considered the nature of
placental endocrinology and its control (Albrecht and Pepe,
1990; Evain-Brion et al., 1990; Masuhiro et al., 1991; Shi
∗ Tel.: +44 20 7594 2133; fax: +44 20 7594 2154. and Zhuang, 1993a,b; Stephanou and Handwerger, 1994;
E-mail address: mark.sullivan@imperial.ac.uk (M.H.F. Sullivan). Petraglia et al., 1995, 1996, 1998; Morrish et al., 1998; Reis

0303-7207/$ – see front matter © 2004 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.mce.2003.03.001
104 M.H.F. Sullivan / Molecular and Cellular Endocrinology 228 (2004) 103–119

Table 1
Endocrine factors, endocrine regulators, and their production by the placenta
Hypothalamic products
GnRH VCT, SYNT Increased in maternal blood (Sorem et al., 1996)
CRH SYNT, VCT, CHOR Increased in maternal blood (Sorem et al., 1996)
Somatostatin VCT Increased in maternal blood (Goldstein et al., 1995)
GHRH No change in pregnancy (Mazlan et al., 1990)
Ghrelin VCT (Gualillo et al., 2001) Decreased in maternal blood (Makino et al., 2002)
TRH SYNT (Bajoria and Babawale, 1998) Increased in maternal blood (Amino et al., 1981)
Dopamine Decreased in maternal blood (Wang et al., 1999)
NPY VCT, CHOR Increased in maternal blood (Petraglia et al., 1989, 1993)
Pituitary products and homologues
Chorionic gonadatrophin SYNT Increased in maternal blood
Placental lactogen SYNT Increased in maternal blood
ACTH SYNT, CHOR Increased in maternal blood (Goland et al., 1992)
␤-Endorphin SYNT, CHOR Increased in maternal blood
␣-MSH SYNT, CHOR No change
Oxytocin SYNT, CHOR Increased in maternal blood
GH SYNT Increased in maternal blood (Mazlan et al., 1990)
Variant GH SYNT (Scippo et al., 1993) Increased in maternal blood (Eriksson, 1989; Frankenne et al., 1987, 1988)
Adrenal products
Glucocorticoids
Deoxycorticosterone Not produced Increased in maternal blood (Dorr et al., 1989; Goland et al., 1992)
Cortisol Not produced Increased in maternal blood (Lockwood et al., 1996)
Mineralocorticoids
Aldosterone Not produced Increased in maternal blood (Dorr et al., 1989)
Gonadal products
Progesterone SYNT, VCT, CHOR Increased in maternal blood
Estrogen SYNT Increased in maternal blood
Activins SYNT, VCT, CHOR Increased in maternal blood
Inhibins SYNT, VCT, CHOR Increased in maternal blood (Qu and Thomas, 1998)
Other local regulators
Interleukins CHOR Increased in maternal blood
Nitric oxide SYNT Increased in maternal blood (Shaamash et al., 2001)
TGF-␤ family SYNT
EGF SYNT, VCT
IGF-I, IGF-II SYNT, VCT, CHOR Increased in maternal blood (IGF-I)
IGFBP-1 Decidua IGFBP-1 increased in maternal blood
Colony-stimulating factor-1 Increased in maternal blood (late) (Tsakonas et al., 1995)
Fibroblast growth factors CYT
CRH-BP SYNT, VCT, CHOR Increased in maternal blood
Extracellular matrix Sanyal and Das (1997)
Hypoxia–HIF-1
The primary source of information is Petraglia et al. (1996), and references therein. Other papers are given to delineate specific data. No localisation
information is shown if this information is not available.

and Petraglia, 2001; Reis et al., 2001; Lacroix et al., 2002; of function is outlined in Fig. 1, showing that the villous
Schneider-Kolsky et al., 2002); the initial part of this review cytotrophoblast are the main source of regulatory factors,
will cover those aspects that are particularly pertinent to un- whereas the syncytiotrophoblast produce the “classic” pep-
derstanding the data described later from cell lines. In ad- tide and steroid hormones (Gaspard et al., 1980; Morrish
dition, thyroid hormones may amplify placental endocrine and Marusyk, 1997). This is not to say that cytotrophoblast
activity in early pregnancy (Maruo et al., 1991), indicat- cannot produce hCG, hPL or steroids in vitro—many stud-
ing that further regulatory pathways may modulate placental ies have shown that they can do so—but rather to emphasise
function. that the main physiological source is the syncytium of intact
placental villi. The functional differentiation must be consid-
ered in all in vitro studies of placental endocrinology. Any
2. The localisation of placental endocrine functions separation of cytotrophoblast from the overlying syncytium
will immediately affect the functions of the syncytium, as
The primary complication in considering placental en- the normal regulation by cytotrophoblast products will be
docrinology is that different placental cell-types have dif- removed. Furthermore there may be regulation in the op-
ferent functions (Dockery et al., 2000). This differentiation posing direction—some studies indicate that chorionic
M.H.F. Sullivan / Molecular and Cellular Endocrinology 228 (2004) 103–119 105

Fig. 1. Cellular sources of the main endocrine factors of human pregnancy in (a) placenta and (b) fetal membranes.

gonadotrophin (syncytial product) regulates progesterone differentiated cells; however, some data indicate that differ-
production (a syncytial and cytotrophoblast function) ent factors may also exert specific regulatory effects on hor-
(Bhattacharyya et al., 1992; Chaudhary et al., 1992); again, mone production (Feinman et al., 1986; Ritvos et al., 1988).
this will be lost on tissue digestion. This type of experimen- With these clear differences in cell biochemistry, it is nec-
tal separation is obligatory if the mechanisms controlling essary to have reliable means of identifying the different pla-
hormone production are to be investigated with precision. cental cell populations, and this must be considered before
The localisation of mRNAs or proteins involved in the con- placental pathways and cell functions can be addressed.
trol of placental endocrine functions has been performed
in cultured explants or intact tissue samples, and of course
provides substantial information on the cellular sources, but 3. Characterisation of different placental cell-types
cannot easily be used to investigate intracellular mecha-
nisms and signal transduction pathways. A complication of The digestion of the human placenta produced a range of
differential functions is that factors implicated in the con- cell-types (Aboagye-Mathiesen et al., 1996a). Gentle tech-
trol of placental endocrinology (see Table 1) may primarily niques can be used to obtain preparations that are composed
influence trophoblast differentiation, which in turn leads to mostly of syncytial trophoblast structures, which can then
changes in endocrinology. For example, factors that increase be cultured. More vigorous techniques produce mixed cell
the formation of syncytia (e.g. EGF) (Morrish et al., 1987, populations, which include villous cytotrophoblast, mononu-
1997) will secondarily increase hCG and hPL production, clear syncytial fragments (which may superficially resemble
as these peptide hormones are produced at higher levels by cytotrophoblast cells) (Huppertz et al., 1999), stromal and
106 M.H.F. Sullivan / Molecular and Cellular Endocrinology 228 (2004) 103–119

endothelial cells. This applies equally to first trimester and membranes provide a larger area of interface with the mater-
to term preparations, as well as to chorion cells from fetal nal tissues than does the placenta; as cervix and myometrium
membranes, which may be contaminated with decidual or are integral to the process of parturition, the regulation of
amnion cells. Extravillous trophoblast may also be obtained their activity by chorionic products may be important in the
from first trimester tissues (Tarrade et al., 2001). continuation of human pregnancy.
Purified cells must be used if the data is to inspire confi-
dence that the data truly pertain to trophoblast, but this may
further remove the cells from their in vivo function which 4. The endocrinology of primary cells from the human
may be controlled by the surrounding cells and matrix. Pla- placenta
cental cell preparations may be crudely assessed by con-
firming the output of expected hormones (e.g. progesterone, It is necessary to review the information obtained from
hCG), but more specific markers are needed to purify tro- isolated human primary placental and trophoblast cell prepa-
phoblast cells. Cytokeratin-7 seems to be expressed by all rations, so that the data from cell lines can be placed in the
trophoblast, and few other cells within the pregnant uterus correct context. These primary cultured cells, and villous
(Potgens et al., 2001). This is an intracellular protein, so it explants, have been used to generate a large body of data on
is not of use in selecting viable cells, but it seems to be a the regulation of placental endocrine functions. Primary tro-
valid way to assess the overall purity of cell preparations. phoblast cells have generally been prepared from two major
The selection of viable cells has been considered in a sources, namely first trimester termination tissue, and term
number of studies and summarised in Workshop Reports placenta or fetal membranes after delivery by elective Cae-
(Frank et al., 2000a, 2001; Guilbert et al., 2002; Morrish sarean section or after normal labour. First trimester samples
et al., 2002). The overall conclusion is that villous cytotro- generally consist of villous and extravillous cytotrophoblast,
phoblast do not express any HLA class-I markers (Frank et which remain as individual cells during culture, whereas
al., 2000a, 2001), so these cells can be purified by removal term tissues can be differentially processed to produce syn-
of all other cell-types by pan-HLA antibodies. Such cells cytial structures or villous cytotrophoblast. The latter may
are also CD9-negative, so depletion of CD9+ cells may also fuse during in vitro culture to form syncytial homologues.
purify villous trophoblast (Yui et al., 1994). The main con- They produce increased quantities of hCG and hPL (Kliman
clusion is that careful characterisation of primary cell prepa- et al., 1986) in culture without additional stimuli, which par-
rations is desirable if reliable data are to be obtained. This allels the in vivo situation—syncytia express higher levels of
is critical in the production of cell lines from primary cells, these peptide hormones than the underlying cytotrophoblast
as mixed cell starting populations may give rise to heteroge- (Morrish and Marusyk, 1997).
neous cell lines, and heterogeneous data. Gestational age will A variety of methods have been used to deal with
also have an impact on the data obtained, as first trimester the problem of contamination of cell preparations by
cytotrophoblast do not form syncytia in vitro, whereas third non-trophoblast components from the villous core. The
trimester cytotrophoblast do, and the reasons for this dif- most robust of these combine immunomagnetic bead selec-
ference in differentiation of these apparently similar villous tion with density gradient separation. The first stage may
cells are not understood. be positive selection (e.g. CD9+ for extravillous cells) or
Villous trophoblast are not the only possible type of negative (e.g. pan-HLA class-I removal of all cells that are
cells that may be appropriate for endocrine studies. There not villous cytotrophoblast). The second step is particularly
are many different cell-types within the trophectoderm- useful for removing cell debris that may accompany neg-
trophoblast lineage, but it seems likely that only two of these ative selection procedures, but may be used to clean up all
are likely to have a major impact throughout pregnancy as cell preparations. Most preparations are >95% CK7 posi-
they are present in significant numbers. Extravillous cells tive, and can be used for a variety of in vitro investigations.
within the placental bed and anchoring villi are CK7 posi- A large number of regulatory interactions between the
tive, but also express HLAG and CD9, which can be used factors produced within the placenta have been identi-
for definitive selection (Tarrade et al., 2001; Hirano et al., fied. These are summarised in Fig. 2, which shows that
1999a). These cells are in immediate contact with maternal most data is available for relatively few of the endocrine
cells, and may therefore play a critical role in regulating factors—primarily GnRH and CRH (hypothalamic-type),
the feto-maternal interface, and preparations of this type hCG, hPL, ␤-endorphin, oxytocin and vGH (pituitary-type),
have been used to study aspects of trophoblast motility or and progesterone, activins and inhibins (gonadal-type).
invasion. There is obviously extensive cross-talk between the factors
The chorion is also CK7 positive (Fig. 1b) and HLAG produced within the placenta, but our knowledge is incom-
positive (Hutter et al., 1996), and can synthesise a range of plete, and the physiology may be more complex than this
endocrine products including progesterone, CRH and hCG diagram indicates. Typical of this may be the control of hCG
(Tonkowicz and Poisner, 1985; Jones et al., 1989; Cooper production by cytokines such as interleukin-1␤ (IL-1␤) and
et al., 1994). It is in immediate proximity to the maternal interleukin-6 (IL-6) (Yagel et al., 1989; Nishino et al., 1990).
decidua, and close to the cervix and myometrium. The fetal It has become clear that some of the regulatory pathways do
M.H.F. Sullivan / Molecular and Cellular Endocrinology 228 (2004) 103–119 107

Fig. 2. Summary of the relationships between endocrine factors and regulators in the human placenta. Known stimulatory (→) and inhibitory ( ) effects
are shown. Main sources are: Petraglia et al. (1996), Margioris et al. (1988), Cronier et al. (1999). Groupings are ‘hypothalamic’ ( ), ‘pituitary’ ( ),
‘gonadal’ ( ) and ‘other’ ( ). (For interpretation of the references to color in this figure legend, the reader is referred to the web version of the article.)

not reflect those in the HPA and HPG axes. In particular, the appropriate substrates, and the determination of estrogenic
control of CRH production from the placenta is stimulated product levels (Wunsch et al., 1986).
by glucocorticoids (King et al., 2001), which may depend The placenta does not express CYP21A2 (21-hydroxy-
on novel interactions between cyclic AMP-dependent and lase), CYP11B1 (11␤-hydroxylase) and CYP11B2 (al-
glucocorticoid-regulated pathways, and the tissue-specific dosterone synthase), and cannot therefore produce any
expression of transcription factors (Cheng et al., 2000; King corticosteroids or aldosterone (Fig. 3). The increased levels
et al., 2002). In addition, some data are contradictory, in that of these steroids found in pregnancy may be derived either
both progesterone (Maruo et al., 1986) and antiprogestin de novo from maternal biosynthesis, or from the further ma-
(Das and Catt, 1987) may inhibit hCG production. ternal metabolism of placental progesterone. The placenta
The placenta also contains steroidogenic enzymes expresses enzymes that can metabolise glucocorticoids,
(Fig. 3), but the placenta cannot synthesise all the primary most notably type-2 11␤-hydroxysteroid dehydrogenase
steroid hormones of pregnancy. The primary biosynthetic (11␤-HSD) (Stewart et al., 1995); this enzyme may be stim-
pathway, including cholesterol recruitment from LDL, and ulated by cyclic AMP (Sun et al., 1998), and down-regulated
cytochrome P450 side-chain cleavage enzyme (P450scc, by catecholamines (Sarkar et al., 2001) and steroids (Sun
CYP11A1) is present (Albrecht and Pepe, 1990; Chaudhary et al., 1998). This converts cortisol to the relatively inactive
et al., 1992; Tuckey et al., 1994), leading to the produc- cortisone, and is thought to protect the fetus, and its devel-
tion of pregnenolone. 3␤-Hydroxysteroid dehydrogenase oping HPA axis from the increased levels of glucocorticoids
(3␤-HSD) converts this to progesterone, one of the pri- present in maternal blood during pregnancy. There is there-
mary steroids of pregnancy. The placenta lacks the enzyme fore a placental–maternal system which parallels adrenal
17␣-hydroxylase/17, 20 lyase (CYP17), and thus cannot steroid production, as well as a placental–fetal system which
further metabolise the progesterone to androgens or es- parallels gonadal steroidogenesis (Fig. 3). In contrast, the
trogens. The production of these steroids relies on fetal type-1 11␤-HSD enzyme is differentially regulated (Sun
and maternal production of dehydroepiandrosterone sul- et al., 1997); this applies particularly to an up-regulation by
phate (DHEAS) and fetal 16␣-OH-DHEAS (Fig. 3). These glucocorticoids in vitro (Sun et al., 2002), which implicates
steroids pass to the placenta, where they are converted to this enzyme in labour rather than pregnancy maintenance.
17␤-estradiol and estriol, respectively. This complex in- These interactions do not provide any information on the
teraction is shown in Fig. 3, and serves to emphasise the intracellular mechanisms through which these effects may
relationship between maternal, fetal and placental tissues. be mediated, which can also be addressed in primary cell
Placental pregnenolone could also be metabolised through or cell line cultures. The main regulatory information from
this route, although the major sources of DHEAS are the primary placental cells in vitro is summarised in Table 2.
adrenals (Fig. 3). The placenta also interconverts estrogens The most general finding is that membrane-permeant cyclic
through the action of 17␤-hydroxysteroid dehydrogenase AMP analogues (dibutyryl (db)- or 8-bromo-derivatives)
(17␤-HSD, CYP19). In vitro studies of aromatase activity increase the output of hCG and of progesterone from these
in cultured trophoblast normally involve the addition of cultured cell preparations. This has been reported in many
108 M.H.F. Sullivan / Molecular and Cellular Endocrinology 228 (2004) 103–119

Fig. 3. The primary steroidogenic pathways of human pregnancy are outlined. The main tissues involved are the placenta ( ), fetal adrenals ( ), fetal liver
( ) and maternal adrenals ( ). Mechanisms and enzymes involved are shown in red. HSD: hydroxysteroid dehydrogenase; S: DHEA sulphotransferase;
16␣OH: 16␣-hydroxylase; S-Sul: steroid sulphatase; CYP11A1: P450 side-chain cleavage enzyme; CYP11B1: P450 11␤-hydroxylase; CYP11B2: P450
aldosterone synthase; CYP17: 17␣-hydroxylase/17, 20 lyase; CYP19: P450 aromatase; CYP21A2: P450 21-hydroxylase (Salido et al., 1990). (For
interpretation of the references to color in this figure legend, the reader is referred to the web version of the article.)

studies (e.g. Feinman et al., 1986; Zhou et al., 1987; Nulsen mal trophoblast function. Such stimulation of progesterone
et al., 1989; Ringler et al., 1989; Dodeur et al., 1990; production clearly parallels the role of cyclic AMP in other
Strauss et al., 1992; Seki et al., 1997; Matsumoto et al., steroidogenic tissues (ovary, adrenal, testis), and suggests
1998), and may clearly be taken as representative of nor- that similar mechanisms may be involved. This may be an

Table 2
Summary of trophoblast regulation
Regulator Target Nature of effect

Cyclic AMP Progesterone Positive (Feinman et al., 1986)


3␤-HSD Positive (Mason et al., 1993)
P450scc (CYP11A1) Positive (Hum and Miller, 1993)
HCG Positive (Feinman et al., 1986)
TGF-␤1 Positive (Ritvos and Eramaa, 1991)
Calcium Progesterone Negative (Zosmer et al., 1997)
HPL Positive (Zeitler et al., 1983)
Inhibin, hCG Positive (Keelan et al., 1994)
PLA2 and melittin hPL Positive (Zeitler et al., 1983, 1991)
Arachidonic acid hPL Positive (Handwerger et al., 1981)
hPL Positive (Zeitler and Handwerger, 1985)
Dexamethasone Inhibin, hCG Positive (Keelan et al., 1994)
CRH No effect (Siler-Khodr et al., 1997)
Norepinephrine hCG, progesterone Positive (Shi and Zhuang, 1993a,b)
Phorbol ester Activin, hCG Positive (Keelan et al., 1994)
HPL Positive (Harman et al., 1986)
M.H.F. Sullivan / Molecular and Cellular Endocrinology 228 (2004) 103–119 109

over-simplification, as one study indicates that the effects of syncytialisation, whereas the type-2 enzyme is mainly cy-
dibutyryl cyclic AMP on progesterone production are more totrophoblastic and decreases in vitro; these findings suggest
variable (Zosmer et al., 1997). The greatest effects of db that these isoforms are differentially regulated (Beaudoin
cyclic AMP were observed when it was added to the cells et al., 1995).
immediately the cultures were set up; a delay of 24 h sub-
stantially decreased the impact of db cyclic AMP on proges-
terone output, and this decrease in steroidogenic capacity has 5. Cell lines from human placenta
been identified previously (Ferguson et al., 1986). Further-
more, the sustained addition of relatively low concentrations There are three main types of cell lines derived from
of db cyclic AMP maintained progesterone production for the human placenta; those which have arisen spontaneously
up to 96 h in vitro; only at higher concentrations of stimulus from cultured cytotrophoblast in vitro, those which have
could any evidence of desensitisation be obtained (Zosmer been immortalised by in vitro transfection with viral genes,
et al., 1997). In other steroidogenic cells, desensitisation may and those from spontaneous choriocarcinomas, which have
be found within a few hours (Sen et al., 1979; Rani et al., been maintained in vitro.
1983; Lefevre et al., 1985). This major difference can easily While many of these cell lines have been used to inves-
be related to the different functions of the product steroids. tigate the control of endocrine functions, there has recently
In the testis and ovary, steroids are produced to (a) develop been some debate about the precise nature of these cells. The
and maintain sexual differentiation and (b) to allow the primary problem seems to be that of defining trophoblast
development of gametes. In both cases these are under the cell-types—for example, many of the choriocarcinoma cell
primary pulsatile control of pituitary products (luteinising lines have been defined as such on the basis of their tissue
hormone and follicle-stimulating hormone) as well as more origin, and by their production of hCG, hPL or steroids. It
local regulators, which leads to a pulsatile hormone release. is now clear that other, non-trophoblast, tumour cells may
Adrenal glucocorticoid production must also be dynamically produce hCG (Duffy, 2001), so that these definitions may
responsive to the body’s requirements. In contrast, proges- not be entirely secure. Ideally, cell lines from the human pla-
terone production in pregnancy must be maintained for 9 centa should be cytokeratin-7 positive (trophoblastic), and
months—it is well established that oophorectomy in the either HLA-negative (villous) or HLAG and CD9 positive
first 2 months of pregnancy (Diczfalusy and Borell, 1961), (extra-villous). Summaries of the characteristics of the main
or administration of progesterone receptor antagonists (e.g. trophoblast cell lines has been published (King et al., 2000;
RU486) (Baulieu, 1985; DeHart and Morehead, 2001) lead Shiverick et al., 2001), which serve mainly to emphasise the
to loss of the conceptus. It should thus be no surprise that lack of rigour that has been applied to the characterisation
placental progesterone production is not readily desensitised of many cell lines. The data from cell lines should be con-
in the presence of stimulus, but rather is sustained. sidered with this proviso in mind.
The nature of the physiological stimulus for placental
progesterone production is not entirely clear. Some in vitro 5.1. Spontaneous cell lines
findings implicate hCG (Shi et al., 1991), but local factors
including GnRH (Shi et al., 1991) and IL-1␤ (Seki et al., A series of primary cell lines have been described, and
1997) may also be involved. Clearly at least one physiologi- these are listed in Table 3. Most of these are spontaneous
cal regulator of placental progesterone production increases transformants, and can be cultured for very many passages.
intracellular cyclic AMP levels, and is itself a placental prod- They may not have been completely characterised, but they
uct; this implicates hCG, but more work is needed to fully all are of placental (or chorionic villous) origin, and may
identify how placental progesterone production is controlled. therefore reflect villous trophoblast function. The character-
The mechanisms have been explored in a series of studies, isation of these cell lines has included some assessment of
which have shown that the uptake and utilisation of LDL endocrine functions (peptide or steroid hormone production)
(Chaudhary et al., 1992), cholesterol transport (Strauss et al., (Table 3), but few studies have used them to investigate the
2000) and expression of CYP11A1 (P450scc) (Chaudhary control mechanisms involved. They have been used exten-
et al., 1992; Tuckey et al., 1994; Hum et al., 1995; sively to investigate other aspects of trophoblast function,
Yamamoto et al., 1994, 1995; Babischkin et al., 1997; including migration and invasion. HT and TL cells both ex-
Beaudoin et al., 1997a) are upreglated during trophoblast pressed estrogen receptor (Ho et al., 1998), but the addition
differentiation and by cyclic AMP analogues. The IGFs of tamoxifen was without effect on cell numbers, which was
can also increase CYP11A1 activity (Nestler, 1987, 1990), in contrast to an antiproliferative effect on BeWo cells. The
while inhibiting aromatase (CYP19) activity (Nestler, 1990, biosynthesis of progesterone by NPC cells was inhibited
1993a). IL-1␤ stimulates CYP19 activity under similar con- by TGF-␤1 (Luo et al., 2002), resulting from a decrease in
ditions (Nestler, 1993b). 17␤-HSD type-1 (which mainly cholesterol transport to mitochondria. They have not been
converts estrone to the more active 17␤-estradiol) is ex- used for any mechanistic studies of placental endocrinology,
pressed primarily in syncytium (Beaudoin et al., 1995; but rather to examine other aspects of trophoblast function,
Bonenfant et al., 2000), and is up-regulated during in vitro including invasion, migration and immunology.
110 M.H.F. Sullivan / Molecular and Cellular Endocrinology 228 (2004) 103–119

Table 3
Summary of data obtained from spontaneous cell lines
TL Ho et al. (1987) CG− PL−
NHT Nagami et al. (1991) CG+ PL−
ED77 Diss et al. (1992) CG+ PL+
HT Ho et al. (1994) CG+ PL−
NPC Rong-Hao et al. (1996) CG+ PL?
ED27 Morgan et al. (1998) CG+ PL+
ED31 Morgan et al. (1998) CG+ PL+
Extravillous primary lines
HTR-8 Graham et al. (1993) Not immortal: 12–15 passages CG− PL+
HT-116 Zdravkovic et al. (1999) Not immortal: 12–15 passages CG? PL?
CG: chorionic gonadotrophin; PL: placental lactogen; (+) positive; (−) negative; (?) not tested.

Two lines with limited life-spans have been described extravillous phenotypes (CK7+/HLAG+/CD9+) are lack-
(Table 3). These cells are grown from villous explants, ing. Most of these cell lines are therefore potential mod-
and have been much more fully characterised as ex- els for investigating the control of endocrine functions, but
pressing an extravillous phenotype (Graham et al., 1993; generally this has not proceeded much beyond demonstra-
Aboagye-Mathiesen et al., 1996b; Zdravkovic et al., 1999). tion of peptide hormone production as part of the charac-
Both can be cultured for 12–15 passages, and HTR-8 cells terisation (e.g. Logan et al., 1992, using SPA-26 cells). The
in particular have been used extensively to investigate the methods used (viral genes and transfection techniques) are
functions of this cell population. These have concentrated summarised in Table 5, and show there is a broad similar-
on the expected in vivo functions of these cells in human ity between the methods used. The precise place these cells
pregnancy, including invasion, apoptosis, proliferation or occupy in trophoblast lineages has been the subject of some
survival, cytokine production and cell–matrix interactions debate (Choy et al., 2000; King et al., 2000; Manyonda et al.,
rather than any possible endocrine functions. NTR-8 cells 2001).
have also been used as the parent cells for the generation of We have investigated the control of hormone production
a transformed cell line (NTR-8/SV40neo) (Graham et al., in TCL-1 cells (Seki et al., 1997), and found that db cyclic
1993), which will be considered in more detail in the next AMP increased progesterone production from these cells.
section of the review. IL-1␤ had similar effects on progesterone production, and
Within the last 18 months, it has become apparent that increased cyclic AMP levels in parallel, implying that there
that one of these cell lines (ED27) is not a reliable model. It might be a functional link. HCG was produced by TCL-1
appears that this cell line became accidentally contaminated cells, but none of the factors tested had any effect on its
with the WISH cell line, which has itself been contaminated production. We attributed this to the insertion of an SV40
by HeLa cells. WISH was originally amniotic epithelium sequence into the promoter region of an hCG subunit, thus
(Hayflick, 1961), but now seems to be genetically indis- damaging the regulatory pathways but permitting a consti-
tinguishable from HeLa, and this also applies ED27 cells tutive synthesis. This emphasises one of the problems with
(Kniss et al., 2002). There are notable phenotypic differ- transfected cells, that alterations to the genome may affect
ences between the cell preparations, which have not yet been endocrine functions and render them less useful models for
elucidated. It is not clear when the contamination occurred, functional studies.
but it must be pointed out that the ED27 cell line still ex-
presses a number of markers and biochemical features that 5.3. Choriocarcinoma cells
are considered typical of human trophoblast (Kniss et al.,
2002). These cells have been used to investigate a number The main choriocarcinoma cell lines are listed in Table 6.
of trophoblast functions (e.g. Kniss et al., 2001), but not The great majority of work has been performed on the old-
specifically endocrine events, so these complications do est cell lines, namely BeWo, JAR (Jar or JAr) (Pattillo and
not impact immediately on our understanding of placental Gey, 1968; Pattillo et al., 1971) and JEG (JEG-3) (Kohler
endocrinology. and Bridson, 1971). These choriocarcinoma cells have been
available for over 30 years, and a correspondingly large lit-
5.2. Transfected cells erature is available. Their characterisation depends partly
on their known origin (in vivo choriocarinoma), and partly
A series of cell lines that have been generated by specific on their production of placental-type hormones (hCG, pro-
transfection with viral genes are listed in Table 4. All these gesterone). In addition, BeWo cells can express mRNA for
cell lines are positive for at least one of hCG or hPL, which is HLAG (Risk and Johnson, 1990) and CD9 protein (Hirano
consistent with trophoblastic origin, but more detailed char- et al., 1999b), which is indicative on an extravillous pheno-
acterisation to identify villous (CK7+/HLA-I−/CD9−) or type. These degree of characterisation may not be considered
M.H.F. Sullivan / Molecular and Cellular Endocrinology 228 (2004) 103–119 111

Table 4
Summary characterisation of transfected cell lines
SPA-26 Chou (1978a,b) CG+ PL− HLAG? CK7?
Primary first trimester cells
HP-W1 Lei et al. (1992b) CG+ PL? HLAGm+ CK7?
Primary term placental cells
HP-A1 Lei et al. (1992b) CG+ PL? HLAGm+ CK7?
Primary term placental cells
HP-A2 Lei et al., 1992b CG+ PL? HLAGm+ CK7?
Primary term placental cells
HTR-8/SV40neo Graham et al. (1993) CG+ PL− HLAG? CK7?
HTR-8 cell line CG- PL+ HLAGic+ CK7?
TCL-1 Lewis et al. (1996) CG+ PL? HLAG− CK7+
Primary term chorion cells CG? PL? HLAG+ CK7+
IST-1 Shih et al., 1998 CG− PL+ HLAG− CK7+
Primary first trimester explants
SGHPL-4 Choy and Manyonda (1998) CG+ PL+ HLAG? CK7?
Primary first trimester cells
SGHPL-5 Choy and Manyonda (1998) CG+ PL+ HLAG? CK7+
Primary first trimester cells
RSVT-2 Khoo et al. (1998) CG− PL+ HLAG? CK7+
HTR-8 cell line CG− PL+ HLAGic+ CK7?
RSVT-2/C Khoo et al. (1998) CG− PL+ HLAG? CK7+
HTR-8 cell line CG− PL+ HLAGic+ CK7?
The source and characterisation where available of parental cell for these cell lines is given to allow comparisons. CG: chorionic gonadotrophin; PL:
placental lactogen; HLAG: human leukocyte antigen-G; CK7: cytokeratin-7; (+) positive; (−) negative; (?) not tested; (m+) mRNA positive; (ic+)
intracellular protein positive.

definitive for trophoblast cells, given the earlier considera- Five cell lines have been generated from combinations
tion of these matters, so all data obtained should be taken of JEG-3 mutants and chorion leave cells, using the meth-
in this context. There are also a small number of studies ods described by Frank et al. (2000b). These cell lines have
in which choriocarcinoma tissue has been studied, showing been deposited at the Deutsche Sammlung von Mikroorgan-
that expression of the LH/hCG receptor is increased in such ismen und Zellkulturen GmbH (DSMZ; German Collection
cells (Lei et al., 1992a). of Microorganisms and Cell Cultures), and more informa-
tion is available through http://www.dsmz.de. They are all
Table 5
cytokeratin+; AC-1M32 is GFAP+, vimentin−, whereas the
Methods to generate transfected cell lines other four cell lines display the opposite phenotype. It is not
known whether the JEG mutants and hybrids are endocrino-
Sequences from SV40
TCL-1 Recombinant retrovirus ZipSV40-6—infection logically active.
HTR-8/SV40neo pSV40neo with large T early region In some studies two or three of the established choriocar-
only—electroporation cinoma cell lines are compared to gain insight into how nor-
SGHPL-4 pSV40neo with large T antigen—poly-l-ornithine mal trophoblast cells might function, and these are outlined
SGHPL-5 pSV40neo with large T antigen—poly-l-ornithine
in Table 7. In general the cells behave very similarly (e.g.
SPA-26 tsA255 temperature sensitive mutant—infection
Bahn et al., 1981; Taylor et al., 1997) suggesting that these
Sequences from other viruses
are at least consistent models for these endpoints. In some
HP-W1 Adenovirus (ori-)SV40 wild type—infection
HP-A1 Adenovirus (ori-)SV40 A209 temperature sensitive studies, cell line specific differences were observed. This is
mutant—infection clearly shown in the work of Mandl et al., 2002 (Table 7);
HP-A2 Adenovirus (ori-)SV40 A209 temperature sensitive none of these studies showed that a factor could stimulate
mutant—infection one cell line, and inhibit the same endpoint in a different cell
IST-1 Recombinant retrovirus coding E6 and E7 proteins
line, indicating that the overall differences were moderate.
from HPV (LXNS16E6E7)—infection
RSVT-2 Plasmid coding for Rous sarcoma In other investigations, more detailed studies have been
virus-T—electroporation performed on one of these cell lines, and these are listed
RSVT-2/C Plasmid coding for Rous sarcoma below. Sometimes these studies included direct compar-
virus-T—electroporation isons with primary trophoblast; these are considered in
112 M.H.F. Sullivan / Molecular and Cellular Endocrinology 228 (2004) 103–119

Table 6
Choriocarcinomas and choriocarcinoma-derived cell lines
BeWo Pattillo and Gey (1968) Choriocarcinoma
JAR Pattillo et al. (1971) Gestational choriocarcinoma
JEG Kohler and Bridson (1971) Choriocarcinoma explants—CK7+
AC1-1 + others Funayama et al. (1997) JEG mutant
ACH1P Frank et al. (2000b) Fusion of AC1-1 (JEG mutant) with term trophoblast
AC-1M32 Frank et al. (2000b)a Fusion of AC1-1 (JEG mutant) with term chorion cells—CK7+
AC-1M46 Frank et al. (2000b)a Fusion of AC1-1 (JEG mutant) with term chorion cells
AC-1M59 Frank et al. (2000b)a Fusion of AC1-1 (JEG mutant) with term chorion cells
AC-1M81 Frank et al. (2000b)a Fusion of AC1-1 (JEG mutant) with term chorion cells
AC-1M88 Frank et al. (2000b)a Fusion of AC1-1 (JEG mutant) with term chorion cells
Only the three initial cell lines are hCG-positive. The AC1 and AC derived cell lines have not been tested.
a The method described in this paper was used to generate these clones, but the clones are not described in detail in the paper.

the next section of the review, in conjunction with the by EGF (Sanyal and Das, 1997). Our unpublished findings
comparisons between different papers which have investi- (Seki et al., 1997) showed that db cyclic AMP increased
gated the same endpoints in choriocarcinoma or primary progesterone production time and dose-dependently.
cells. JEG-3 cells: cAMP analogues increase P450scc (CYP-
BeWo cells: the production of hCG is dependent on cyclic 11A1) by transcription and adrenodoxin post-transcription-
AMP, protein kinase C and calcium (Kaiho et al., 1984; ally (Brentano and Miller, 1992; Moore et al., 1992; Guo
Futamura et al., 1987; Nishino et al., 1991), and is also et al., 1994; Beaudoin et al., 1997b), 3␤-HSD (Beaudoin
increased by LHRH (GnRH) (Kaiho et al., 1984), EGF et al., 1997b), and TGF-␤1 production (Ritvos and Eramaa,
(Futamura et al., 1987), keratinocyte growth factor (Matsui 1991). Calcium decreased CYP11A1 and 3␤-HSD activity
et al., 2000). HCG production (used as a differentiation (Beaudoin et al., 1997b). 17␤-HSD-1 was increased by cal-
marker) was also increased by culture on Matrigel (Hohn cium and cAMP synergistically (Beaudoin et al., 1997b).
et al., 1992, 1996), and by the addition of methotrexate 11␤-HSD was increased by cAMP, but not by protein kinase
(Taylor et al., 1991); the latter increased aromatase activity. C (Pasquarette et al., 1996). Activin A may be an autocrine
11␤-HSD activity was decreased by catecholamines (Sarkar regulator of JEG-3 cell steroidogenesis (Ni et al., 2000),
et al., 2001). Proliferation of BeWo cells was decreased by and TGF-␤1 inhibited progesterone and estradiol production
estradiol or progesterone (Kaiho et al., 1984), and increased (Luo et al., 2002). TNF-␣ increased estrogen and hCG pro-

Table 7
Regulation of choriocarcinoma cells
BeWo JEG-3 JAR

cAMP analogues on hCG production + + nd Chou et al. (1978)


Progesterone produced, no estrogens + + + Bahn et al. (1981)
Retinoic acid on hCG + + + Kato and Braunstein (1991)
Retinoic acid on progsterone + + No Kato and Braunstein (1991)
T3 on vGH family members +++ ++ + Nickel and Cattini (1991)
Insulin on hCG production nd + + Ren and Braunstein (1991)
␤FGF on 17␤-HSD-1 No ++ ++ Lewintre et al. (1994)
VGH and hCS RNA Present Present Present Lytras et al. (1994)
M-CSF on proliferation and hCG No No No Saito et al. (1994)
Benzo[␣]pyrene on hCG production − No nd Zhang et al. (1995)
Nitric oxide on hCG − − nd Myat et al. (1996)
cAMP on CRH promoter activity + + nd Scatena and Adler (1996)
Hypoxia on hCG − − − Strohmer et al. (1997)
Hypoxia on VEGF ++ ++ ++ Taylor et al. (1997)
Hypoxia on hCG +/− +/− +/− Taylor et al. (1997)
Tamoxifen on growth − − − Ho et al. (1998)
Steroid uptake and steroyl-sulphatase Present Present Present Ugele and Simon (1999)
Chemokines on proliferation and hCG + + + Ishii et al. (2000)
Insulin on proliferation No + ++ Mandl et al. (2002)
IGF-I on proliferation No No – Mandl et al. (2002)
Insulin on hCG + + No Mandl et al. (2002)
IGF-I on hCG No + ++ Mandl et al. (2002)
Trialkyltin compounds (endocrine disruptors) on hCG and progesterone ++ ++ ++ Nakanishi et al. (2002)
(+) positive effect; (−) negative effect; no: no effect; nd: not done.
M.H.F. Sullivan / Molecular and Cellular Endocrinology 228 (2004) 103–119 113

duction, and decreased progesterone production (Pedersen transfected human genes. Progesterone inhibits hCG pro-
et al., 1995). duction from transfected Rcho-1 cells (Yamamoto et al.,
Jar cells: hCG production was increased by 8-bromo-ade- 2001). Human CRH promoter is active in BeWo and JEG-3
nosine or db cyclic AMP (Hussa et al., 1975, 1977; Martell cells, but not in Rcho-1 cells; this species difference is
and Ruddon, 1990; Sibley et al., 1991), IL-1␤ and TNF-␣ attributed to the presence of different transcription factors
(Yanushpolsky et al., 1993), GnRH (Ertl et al., 1993), and (Scatena and Adler, 1996). BT-1 trophoblastic cells from
PGE2 (Yagel et al., 1989). This latter paper found no ef- bovine blastocyst express interferon-␶ and bovine placental
fect of IL-1␤ on hCG output (Yagel et al., 1989), which lactogen (Shimada et al., 2001).
differs from the later results of Yanushpolsky et al., 1993.
The reasons for this difference are not clear. Early data sug-
gested that EGF did not affect hCG production from this 7. Summary of studies performed and limitations
cell line (Huot et al., 1981), but recent findings show that
EGF increases hCG production, through a mechanism in- A wide range of endpoints have been investigated using
volving cross-talk with protein kinase C (Baker et al., 1998). the cell lines described in this review. Many of them are
Aromatase activity was also increased by db cyclic AMP of endocrine nature, and have investigated biosynthetic or
(Bellino et al., 1978). Removal of calcium, as well as the metabolic pathways. In general terms the data obtained from
addition of the calcium ionophore A23187, decreased hCG the cell lines, and from primary cells are comparable. This
production (Hussa, 1977). Jar cells interact with primary overall consistency is encouraging, suggesting that, despite
cytotrophoblasts to increase net hCG and hPL production the many different cellular models used, the data do reflect
(Eldar-Geva et al., 1993), but the details of this cross-talk in vitro functions. This is most clearly seen in the production
have not been elucidated. of hormones, particularly hCG and progesterone, which is
up-regulated by cyclic AMP analogues, and is dependent on
the presence of calcium in the media. Protein kinase C also
6. Cell lines from other species seems to be a positive effector in these processes. This most
clearly in comparing Table 2 (primary cells) and Table 7
While there is a significant body of information on en- (choriocarcinoma cell lines). EGF and GnRH also seem to
docrine function in pregnancy in other species, there are few be generic positive factors for hormone production, and for
cell lines of non-human origin. This review will therefore proliferation in the case of EGF.
focus on the endocrinology of the human placenta, and of There are also some clear differences between primary
the cell lines derived from it, although cell lines from other trophoblast and cell lines, as indicated by the comparative
species will be considered if they provide particular insights. studies shown in Table 8, but there are generally fewer dif-
Placental structures may differ substantially between differ- ferences than similarities. It must be pointed out that entirely
ent species, so direct comparisons between apparently re- negative findings (e.g. M-CSF does not affect the chorio-
lated cell-types may not be entirely valid. carcinoma cells, Saito et al., 1994) are usually published in
Rcho-1 cells (Faria and Soares, 1991) were developed context of positive data (M-CSF affects primary cells); there
as a model to study rat trophoblast function, including may therefore be more information available than has been
placental lactogen synthesis. They have also been used to published, showing more differences between the cell lines
investigate endocrine functions, including the activity of than the generally clear findings summarised in Fig. 2.

Table 8
Comparison of primary and cell line data
Similar findings
Hussa (1977), Hussa et al. (1977): calcium and cyclic AMP required for hCG production from JAR and primary cells
Bahn et al. (1981): all three choriocarcinomas resemble primary cells in the steroidogenic enzymes present
Pedersen et al. (1995): TNF-␣ has similar effects on primary and JEG-3 cells hormone production
Ritvos and Eramaa (1991): JEG-3 and primary trophoblast TGF-␤1 increased by cAMP
Sibley et al. (1991): 8Br-adenosine increased hCG production from JAR and cytotrohoblast cells
Stephanou and Handwerger (1995): the effects of retinoic acid and thyroid hormone and similar in cytotrophoblast, and in transfected BeWo cells
Conrad et al. (1996): expression of erythorpoietin similar in primary and JAR cells
Seki et al. (1997): TCL-1 cell line and primary cells respond similarly to db cyclic AMP
Sarkar et al. (2001): primary and BeWo 11␤-HSD decreased by catecholamines
Different findings
Hoshina et al. (1985): cytotrophoblast and choriocarcinoma show different patterns of HCG and hPL expression
Maruo et al. (1986): in primary cells progesterone inhibited hCG production, but was without effect in choriocarcinoma cells
Ren and Braunstein (1991): insulin increased hCG production from JEG-3 and JAR cells, but not primary cytotrophoblast
Saito et al. (1994): M-CSF induces proliferation and differentiation of normal trophoblast, but not choriocarcinoma cells
Seki et al. (1997): different responses of primary and TCL-1 cells to IL-1␤
114 M.H.F. Sullivan / Molecular and Cellular Endocrinology 228 (2004) 103–119

The greatest limitation from an endocrine perspective is Bajoria, R., Babawale, M., 1998. Ontogeny of endogenous secretion of
that referred to earlier, that the cellular heterogeneity of pla- immunoreactive-thyrotropin releasing hormone by the human placenta.
J. Clin. Endocrinol. Metab. 83, 4148–4155.
cental trophoblast is integral to the normal physiology of Baker, V.L., Murai, J.T., Taylor, R.N., 1998. Downregulation of protein ki-
this organ, and cell lines will only reflect a part of this. nase C by phorbol ester increases expression of epidermal growth fac-
tor receptors in transformed trophoblasts and amplifies human chori-
onic gonadotropin production. Placenta 19, 475–482.
Baulieu, E.E., 1985. Contragestion by antiprogestin: a new approach to
8. Other trophoblast functions human fertility control. Ciba Found. Symp. 115, 192–210.
Beaudoin, C., Blomquist, C.H., Tremblay, Y., 1995. Gene expression of 17
Placental trophoblast also exhibit a range of non-endocrine beta-hydroxysteroid dehydrogenase type 2 isozyme in primary cultures
functions, including cell motility, invasion (Kliman and of human trophoblasts predicts different mechanisms regulating type
Feinberg, 1990), homotypic fusion (formation of syncy- 1 and type 2 enzymes. Endocrinology 136, 3807–3814.
Beaudoin, C., Blomquist, C.H., Bonenfant, M., Tremblay, Y., 1997a. Ex-
tia), complex cell–cell and cell–matrix interactions, and pression of the genes for 3 beta-hydroxysteroid dehydrogenase type
apoptosis. These may be under the control of factors that 1 and cytochrome P450scc during syncytium formation by human
are integral parts of the endocrine axes (e.g. progesterone placental cytotrophoblast cells in culture and the regulation by pro-
inhibits matrix metalloproteinase production; Shimonovitz gesterone and estradiol. J. Endocrinol. 154, 379–387.
et al., 1998), or regulate the endocrine axes (e.g. nitric Beaudoin, C., Bonenfant, M., Tremblay, Y., 1997b. Regulation of cy-
tochrome P450 cholesterol side-chain cleavage, 3 beta-hydroxysteroid
oxide). While these aspects lie beyond the scope of this dehydrogenase/delta 5-delta 4 isomerase type 1 and estradiol-17 beta-
review, the attention of readers is directed to other reviews hydroxysteroid dehydrogenase mRNA levels by calcium in human
and papers that consider these aspects of trophoblast func- choriocarcinoma JEG-3 cells. Mol. Cell. Endocrinol. 133, 63–71.
tion in more detail. It is possible that such processes may Bellino, F.L., Hussa, R.O., Osawa, Y., 1978. Estrogen synthetase in chori-
be modelled accurately by cell lines, as they normally in- ocarcinoma cell culture. Stimulation by dibutyryl cyclic adenosine
monophosphate and theophylline. Steroids 32, 37–44.
volve isolated cytotrophoblast rather than combinations of Bhattacharyya, S., Chaudhary, J., Das, C., 1992. Antibodies to hCG
different cell-types which seem to be more important in the inhibit progesterone production from human syncytiotrophoblast cells.
endocrine functions considered in this review. Placenta 13, 135–139.
Bonenfant, M., Provost, P.R., Drolet, R., Tremblay, Y., 2000. Localization
of type 1 17 beta-hydroxysteroid dehydrogenase mRNA and protein
in syncytiotrophoblasts and invasive cytotrophoblasts in the human
Acknowledgements term villi. J. Endocrinol. 165, 217–222.
Brentano, S.T., Miller, W.L., 1992. Regulation of human cytochrome
I must thank the colleagues who have helped in the warit- P450scc and adrenodoxin messenger ribonucleic acids in JEG-3 cy-
ing of this review by reading the manuscript or contributing totrophoblast cells. Endocrinology 131, 3010–3018.
Chaudhary, J., Bhattacharyya, S., Das, C., 1992. Regulation of proges-
information and insights that lie beyond the published liter- terone secretion in human syncytiotrophoblast in culture by human
ature. In particularly I thank Drs. Mandy Donaldson, Guy chorionic gonadotropin. J. Steroid Biochem. Mol. Biol. 42, 425–432.
Whitley, Ashley Moffett and Hans-Georg Frank. Cheng, Y.H., Nicholson, R.C., King, B., Chan, E.C., Fitter, J.T., Smith, R.,
2000. Glucocorticoid stimulation of corticotropin-releasing hormone
gene expression requires a cyclic adenosine 3 ,5 -monophosphate reg-
ulatory element in human primary placental cytotrophoblast cells. J.
Clin. Endocrinol. Metab. 85, 1937–1945.
References Chou, J.Y., 1978a. Human placental cells transformed by tsA mutants of
simian virus 40: a model system for the study of placental functions.
Aboagye-Mathiesen, G., Laugesen, J., Zdravkovic, M., Ebbesen, P., 1996a. Proc. Natl. Acad. Sci. U.S.A. 75, 1409–1413.
Isolation and characterization of human placental trophoblast subpopu- Chou, J.Y., 1978b. Establishment of clonal human placental cells synthe-
lations from first-trimester chorionic villi. Clin. Diagn. Lab. Immunol. sizing human choriogonadotropin. Proc. Natl. Acad. Sci. U.S.A. 75,
3, 14–22. 1854–1858.
Aboagye-Mathiesen, G., Zdravkovic, M., Toth, F.D., Graham, C.H., Lala, Chou, J.Y., Wang, S.S., Robinson, J.C., 1978. Regulation of the synthesis
P.K., Ebbesen, P., 1996b. Altered expression of the tumor suppres- of human chorionic gonadotrophin by 5-bromo-2 -deoxyuridine and
sor/oncoprotein p53 in SV40 Tag-transformed human placental tro- dibutyryl cyclic AMP in trophoblastic and nontrophoblastic tumor
phoblast and malignant trophoblast cell lines. Early Pregnancy 2, cells. J. Clin. Endocrinol. Metab. 47, 46–51.
102–112. Choy, M.Y., Manyonda, I.T., 1998. The phagocytic activity of human first
Albrecht, E.D., Pepe, G.J., 1990. Placental steroid hormone biosynthesis trimester extravillous trophoblast. Hum. Reprod. 13, 2941–2949.
in primate pregnancy. Endocrinol. Rev. 11, 124–150. Choy, M.Y., St. Whitley, G., Manyonda, I.T., 2000. Efficient, rapid and
Amino, N., Yamada, T., Mitsuma, T., Nogimori, T., Tanizawa, O., reliable establishment of human trophoblast cell lines using poly-l-
Kawashima, M., Kurachi, K., Miyai, K., 1981. Increase in plasma ornithine. Early Pregnancy 4, 124–143.
thyrotropin-releasing hormone in normal human pregnancy. J. Clin. Conrad, K.P., Benyo, D.F., Westerhausen-Larsen, A., Miles, T.M., 1996.
Endocrinol. Metab. 53, 1288–1290. Expression of erythropoietin by the human placenta. FASEB J. 10,
Babischkin, J.S., Grimes, R.W., Pepe, G.J., Albrecht, E.D., 1997. Estrogen 760–768.
stimulation of P450 cholesterol side-chain cleavage activity in cultures Cooper, E.S., Brooks, A.N., Miller, M.R., Greer, I.A., 1994. Corti-
of human placental syncytiotrophoblasts. Biol. Reprod. 56, 272–278. cotrophin-releasing factor immunostaining is present in placenta and
Bahn, R.S., Worsham, A., Speeg Jr., K.V., Ascoli, M., Rabin, D., 1981. fetal membranes from the first trimester onwards and is not affected
Characterization of steroid production in cultured human choriocarci- by labour or administration of mifepristone. Clin. Endocrinol. (Ox-
noma cells. J. Clin. Endocrinol. Metab. 52, 447–450. ford) 41, 677–683.
M.H.F. Sullivan / Molecular and Cellular Endocrinology 228 (2004) 103–119 115

Cronier, L., Guibourdenche, J., Niger, C., Malassine, A., 1999. Oestra- women and partial characterization of the placental GH variant. J.
diol stimulates morphological and functional differentiation of human Clin. Endocrinol. Metab. 66, 1171–1180.
villous cytotrophoblast. Placenta 20, 669–676. Funayama, H., Gaus, G., Ebeling, I., Takayama, M., Füzesi, L., Huppertz,
Das, C., Catt, K.J., 1987. Antifertility actions of the progesterone antago- B., Kaufmann, P., Frank, H.G., 1997. Parent cells for trophoblast
nist RU 486 include direct inhibition of placental hormone secretion. hybridization. II. ACI and related trophoblast cell lines, a family
Lancet 2, 599–601. of HGPRT-negative mutants of the choriocarcinoma cell line JEG-3.
DeHart, R.M., Morehead, M.S., 2001. Mifepristone. Ann. Pharmacother. Troph. Res. 10, 191–201.
35, 707–719. Futamura, K., Maruo, T., Mochizuki, M., 1987. Differential effects of
Diczfalusy, E., Borell, U., 1961. Influence of oophorectomy on steroid dibutyryl cyclic AMP and epidermal growth factor on the synthe-
excretion in early pregnancy. J. Clin. Endocrinol. 21, 1119–1126. sis and secretion of human chorionic gonadotropin and its subunits
Diss, E.M., Gabbe, S.G., Moore, J.W., Kniss, D.A., 1992. Study of by trophoblastic and non-trophoblastic cells. Nippon Sanka Fujinka
thromboxane and prostacyclin metabolism in an in vitro model of Gakkai Zasshi 39, 1641–1648.
first-trimester human trophoblast. Am. J. Obstet. Gynecol. 167, 1046– Gaspard, U.J., Hustin, J., Reuter, A.M., Lambotte, R., Franchimont, P.,
1052. 1980. Immunofluorescent localization of placental lactogen, chorionic
Dockery, P., Bermingham, J., Jenkins, D., 2000. Structure–function rela- gonadotrophin and its alpha and beta subunits in organ cultures of
tions in the human placenta. Biochem. Soc. Trans. 28, 202–208. human placenta. Placenta 1, 135–144.
Dodeur, M., Malassine, A., Bellet, D., Mensier, A., Evain-Brion, D., 1990. Glinoer, D., de Nayer, P., Bourdoux, P., Lemone, M., Robyn, C., van
Characterization and differentiation of human first trimester placenta Steirteghem, A., Kinthaert, J., Lejeune, B., 1990. Regulation of mater-
trophoblastic cells in culture. Reprod. Nutr. Dev. 30, 183–192. nal thyroid during pregnancy. J. Clin. Endocrinol. Metab. 71, 276–287.
Dorr, H.G., Heller, A., Versmold, H.T., Sippell, W.G., Herrmann, M., Goland, R.S., Conwell, I.M., Warren, W.B., Wardlaw, S.L., 1992. Placental
Bidlingmaier, F., Knorr, D., 1989. Longitudinal study of progestins, corticotropin-releasing hormone and pituitary-adrenal function during
mineralocorticoids, and glucocorticoids throughout human pregnancy. pregnancy. Neuroendocrinology 56, 742–749.
J. Clin. Endocrinol. Metab. 68, 863–868. Goldstein, A., Armony-Sivan, R., Rozin, A., Weller, A., 1995. Somato-
Duffy, M.J., 2001. Clinical uses of tumor markers: a critical review. Crit. statin levels during infancy, pregnancy, and lactation: a review. Pep-
Rev. Clin. Lab. Sci. 38, 225–262. tides 16, 1321–1326.
Eldar-Geva, T., Rachmilewitz, J., de Groot, N., Hochberg, A., 1993. In- Graham, C.H., Hawley, T.S., Hawley, R.G., MacDougall, J.R., Kerbel,
teraction between choriocarcinoma cell line (JAr) and human cytotro- R.S., Khoo, N., Lala, P.K., 1993. Establishment and characteriza-
phoblasts in vitro. Placenta 14, 217–223. tion of first trimester human trophoblast cells with extended lifespan.
Eriksson, L., 1989. Growth hormone in human pregnancy. Maternal 24-h Exp. Cell Res. 206, 204–211.
serum profiles and experimental effects of continuous GH secretion. Gualillo, O., Caminos, J., Blanco, M., Garcia-Caballero, T., Kojima, M.,
Acta Obstet. Gynecol. Scand. Suppl. 147, 1–38. Kangawa, K., Dieguez, C., Casanueva, F., 2001. Ghrelin, a novel
Ertl, T., Torok, A., Bodis, J., Pflieger, S., Zwirner, M., Hanf, V., Tin- placental-derived hormone. Endocrinology 142, 788–794.
neberg, H.R., 1993. Spontaneous and gonadotropin-releasing hormone
Guilbert, L.J., Winkler-Lowen, B., Sherburne, R., Rote, N.S., Li, H.,
induced human chorionic gonadotropin release from superfused JAR
Morrish, D.W., 2002. Preparation and functional characterization of
choriocarcinoma cells. Arch. Gynecol. Obstet. 253, 43–49.
villous cytotrophoblasts free of syncytial fragments. Placenta 23, 175–
Evain-Brion, D., Alsat, E., Mirlesse, V., Dodeur, M., Scippo, M.L., Hen-
183.
nen, G., Frankenne, F., 1990. Regulation of growth hormone secretion
Guo, I.C., Tsai, H.M., Chung, B.C., 1994. Actions of two different
in human trophoblastic cells in culture. Horm. Res. 33, 256–259.
cAMP-responsive sequences and an enhancer of the human CYP11A1
Faria, T.N., Soares, M.J., 1991. Trophoblast cell differentiation: establish-
(P450scc) gene in adrenal Y1 and placental JEG-3 cells. J. Biol.
ment, characterization, and modulation of a rat trophoblast cell line
Chem. 269, 6362–6369.
expressing members of the placental prolactin family. Endocrinology
Handwerger, S., Barrett, J., Barry, S., Markoff, E., Zeitler, P., Cwikel, B.,
129, 2895–2906.
Siegel, M., 1981. Stimulation of human placental lactogen release by
Feinman, M.A., Kliman, H.J., Caltabiano, S., Strauss, J.F., 1986. 8-Bromo-
arachidonic acid. Mol. Pharmacol. 20, 609–613.
3 ,5 -adenosine monophosphate stimulates the endocrine activity of
Harman, I., Zeitler, P., Ganong, B., Bell, R.M., Handwerger, S., 1986.
human cytotrophoblasts in culture. J. Clin. Endocrinol. Metab. 63,
Sn-1,2-diacylglycerols and phorbol esters stimulate the synthesis and
1211–1217.
release of human placental lactogen from placental cells: a role for
Ferguson, K.A., Mitchell, B.F., Tanswell, A.K., 1986. Human chorion cells
protein kinase C. Endocrinology 119, 1239–1244.
respond to growth factors but lose steroidogenic capacity in primary
Hayflick, L., 1961. The establishment of a line (WISH) of human amnion
monolayer cell culture. In Vitro Cell Dev. Biol. 22, 320–324.
cells in continuous cultivation. Exp. Cell Res. 23, 14–20.
Frank, H.G., Genbacev, O., Blaschitz, A., Chen, C.P., Clarson, L., Evain-
Brion, D., Gardner, L., Malek, A., Morrish, D., Loke, Y.W., Tarrade, Hirano, T., Higuchi, T., Ueda, M., Inoue, T., Kataoka, N., Maeda, M.,
A., 2000a. Cell culture models of human trophoblast—primary culture Fujiwara, H., Fujii, S., 1999a. CD9 is expressed in extravillous tro-
of trophoblast—a workshop report. Placenta 21 (Suppl. A), S120–122. phoblasts in association with integrin alpha3 and integrin alpha5. Mol.
Frank, H.G., Gunawan, B., Ebeling-Stark, I., Schulten, H.J., Funayama, H., Hum. Reprod. 5, 162–167.
Cremer, U., Huppertz, B., Gaus, G., Kaufmann, P., Fuzesi, L., 2000b. Hirano, T., Higuchi, T., Katsuragawa, H., Inoue, T., Kataoka, N., Park,
Cytogenetic and DNA-fingerprint characterization of choriocarcinoma K.R., Ueda, M., Maeda, M., Fujiwara, H., Fujii, S., 1999b. CD9 is
cell lines and a trophoblast/choriocarcinoma cell hybrid. Cancer Genet. involved in invasion of human trophoblast-like choriocarcinoma cell
Cytogenet. 116, 16–22. line, BeWo cells. Mol. Hum. Reprod. 5, 168–174.
Frank, H.G., Morrish, D.W., Potgens, A., Genbacev, O., Kumpel, B., Ho, C.K., Chiang, H., Li, S.Y., Yuan, C.C., Ng, H.T., 1987. Establishment
Caniggia, I., 2001. Cell culture models of human trophoblast: primary and characterization of a tumorigenic trophoblast-like cell line from
culture of trophoblast—a workshop report. Placenta 22 (Suppl. A), a human placenta. Cancer Res. 47, 3220–3224.
S107–109. Ho, C.K., Li, S.Y., Yu, K.J., Wang, C.C., Chiang, H., Wang, S.Y., 1994.
Frankenne, F., Rentier-Delrue, F., Scippo, M.L., Martial, J., Hennen, Characterization of a human tumorigenic, poorly differentiated tro-
G., 1987. Expression of the growth hormone variant gene in human phoblast cell line. In Vitro Cell Dev. Biol. Anim. 30A, 415–417.
placenta. J. Clin. Endocrinol. Metab. 64, 635–637. Ho, C.K., Chi, C.W., Yu, K.J., Wang, S.Y., 1998. Tamoxifen-mediated
Frankenne, F., Closset, J., Gomez, F., Scippo, M.L., Smal, J., Hennen, anti-cellular effect against a choriocarcinoma cell line. Int. J. Oncol.
G., 1988. The physiology of growth hormones (GHs) in pregnant 12, 1171–1176.
116 M.H.F. Sullivan / Molecular and Cellular Endocrinology 228 (2004) 103–119

Hohn, H.P., Parker Jr., C.R., Boots, L.R., Denker, H.W., Hook, M., 1992. King, B.R., Smith, R., Nicholson, R.C., 2002. Novel glucocorticoid and
Modulation of differentiation markers in human choriocarcinoma cells cAMP interactions on the CRH gene promoter. Mol. Cell. Endocrinol.
by extracellular matrix: on the role of a three-dimensional matrix 194, 19–28.
structure. Differentiation 51, 61–70. Kliman, H.J., Nestler, J.E., Sermasi, E., Sanger, J.M., Strauss III, J.F.,
Hohn, H.P., Grummer, R., Bosserhoff, S., Graf-Lingnau, S., Reuss, B., 1986. Purification, characterization, and in vitro differentiation of cy-
Backer, C., Denker, H.W., 1996. The role of matrix contact and of totrophoblasts from human term placentae. Endocrinology 118, 1567–
cell–cell interactions in choriocarcinoma cell differentiation. Eur. J. 1582.
Cell Biol. 69, 76–85. Kliman, H.J., Feinberg, R.F., 1990. Human trophoblast–extracellular ma-
Hoshina, M., Boothby, M., Hussa, R., Pattillo, R., Camel, H.M., Boime, trix (ECM) interactions in vitro: ECM thickness modulates morphol-
I., 1985. Linkage of human chorionic gonadotrophin and placental ogy and proteolytic activity. Proc. Natl. Acad. Sci. U.S.A. 87, 3057–
lactogen biosynthesis to trophoblast differentiation and tumorigenesis. 3061.
Placenta 6, 163–172. Kniss, D.A., Rovin, B., Fertel, R.H., Zimmerman, P.D., 2001. Blockade
Hum, D.W., Miller, W.L., 1993. Transcriptional regulation of human genes NF-kappaB activation prohibits TNF-alpha-induced cyclooxygenase-2
for steroidogenic enzymes. Clin. Chem. 39, 333–340. gene expression in ED27 trophoblast-like cells. Placenta 22, 80–89.
Hum, D.W., Aza-Blanc, P., Miller, W.L., 1995. Characterization of pla- Kniss, D.A., Xie, Y., Li, Y., Kumar, S., Linton, E.A., Cohen, P., Fan-
cental transcriptional activation of the human gene for P450scc. DNA Havard, P., Redman, C.W., Sargent, I.L., 2002. ED(27) trophoblast-
Cell Biol. 14, 451–463. like cells isolated from first-trimester chorionic villi are genetically
Huot, R.I., Foidart, J.M., Nardone, R.M., Stromberg, K., 1981. Differential identical to HeLa cells yet exhibit a distinct phenotype. Placenta 23,
modulation of human chorionic gonadotropin secretion by epidermal 32–43.
growth factor in normal and malignant placental cultures. J. Clin. Kohler, P.O., Bridson, W.E., 1971. Isolation of hormone-producing clonal
Endocrinol. Metab. 53, 1059–1063. lines of human choriocarcinoma. J. Clin. Endocrinol. Metab. 32, 683–
Huppertz, B., Frank, H.G., Reister, F., Kingdom, J., Korr, H., Kaufmann, 687.
P., 1999. Apoptosis cascade progresses during turnover of human Lacroix, M.C., Guibourdenche, J., Frendo, J.L., Muller, F., Evain-Brion,
trophoblast: analysis of villous cytotrophoblast and syncytial fragments D., 2002. Human placental growth hormone—a review. Placenta
in vitro. Lab. Invest. 79, 1687–1702. 23 (Suppl. A), S87–94.
Hussa, R.O., 1977. Studies on human chorionic gonadotropin secretion: Lefevre, A., Finaz, C., Berthelon, M.C., Saez, J.M., 1985. Modulation of
effects of EGTA, lanthanum, and the ionophore A23187. J. Clin. cultured mouse Leydig cells adenylate cyclase by forskolin and hCG.
Endocrinol. Metab. 44, 520–529. Mol. Cell. Endocrinol. 40, 107–114.
Hussa, R.O., Story, M.T., Pattillo, R.A., 1975. Regulation of human Lei, Z.M., Rao, C.V., Ackerman, D.M., Day, T.G., 1992a. The expression
chorionic gonadotropin (hCG) secretion by serum and dibutyryl cyclic of human chorionic gonadotropin/human luteinizing hormone recep-
AMP in malignant trophoblast cells in vitro. J. Clin. Endocrinol. tors in human gestational trophoblastic neoplasms. J. Clin. Endocrinol.
Metab. 40, 401–405. Metab. 74, 1236–1241.
Hussa, R.O., Story, M.T., Pattillo, R.A., Kemp, R.G., 1977. Effect of cyclic Lei, K.J., Gluzman, Y., Pan, C.J., Chou, J.Y., 1992b. Immortalization of
3 :5 -AMP derivatives prostaglandins and related agents on human virus-free human placental cells that express tissue-specific functions.
chorionic gonadotropin secretion in human malignant trophoblast in Mol. Endocrinol. 6, 703–712.
culture. In Vitro 13, 443–449. Lewintre, E.J., Orava, M., Vihko, R., 1994. Regulation of 17 beta-
Hutter, H., Hammer, A., Blaschitz, A., Hartmann, M., Ebbesen, P., Dohr, hydroxysteroid dehydrogenase type 1 by epidermal growth factor
G., Ziegler, A., Uchanska-Ziegler, B., 1996. Expression of HLA class and transforming growth factor-alpha in choriocarcinoma cells. En-
I molecules in human first trimester and term placenta trophoblast. docrinology 135, 2629–2634.
Cell Tissue Res. 286, 439–447. Lewis, M.P., Clements, M., Takeda, S., Kirby, P.L., Seki, H., Lonsdale,
Ishii, M., Hayakawa, S., Suzuki, M.K., Yoshino, N., Honda, M., Nishi- L.B., Sullivan, M.H., Elder, M.G., White, J.O., 1996. Partial char-
narita, S., Chishima, F., Nagaishi, M., Satoh, K., 2000. Expression acterization of an immortalized human trophoblast cell-line, TCL-1,
of functional chemokine receptors of human placental cells. Am. J. which possesses a CSF-1 autocrine loop. Placenta 17, 137–146.
Reprod. Immunol. 44, 365–373. Lockwood, C.J., Radunovic, N., Nastic, D., Petkovic, S., Aigner, S.,
Jones, S.A., Brooks, A.N., Challis, J.R., 1989. Steroids modulate Berkowitz, G.S., 1996. Corticotropin-releasing hormone and related
corticotropin-releasing hormone production in human fetal membranes pituitary-adrenal axis hormones in fetal and maternal blood during the
and placenta. J. Clin. Endocrinol. Metab. 68, 825–830. second half of pregnancy. J. Perinat. Med. 24, 243–251.
Kaiho, T., Sekiya, S., Shirotake, S., Takamizawa, H., 1984. Effects of var- Logan, S.K., Fisher, S.J., Damsky, C.H., 1992. Human placental cells
ious agents on the proliferation and hCG secretion of choriocarcinoma transformed with temperature-sensitive simian virus 40 are immortal-
cells in vitro. Nippon Sanka Fujinka Gakkai Zasshi 36, 1801–1806. ized and mimic the phenotype of invasive cytotrophoblasts at both
Kato, Y., Braunstein, G.D., 1991. Retinoic acid stimulates placental hor- permissive and nonpermissive temperatures. Cancer Res. 52, 6001–
mone secretion by choriocarcinoma cell lines in vitro. Endocrinology 6009.
128, 401–407. Luo, S., Yu, H., Wu, D., Peng, C., 2002. Transforming growth factor-
Keelan, J., Song, Y., France, J.T., 1994. Comparative regulation of inhibin, beta1 inhibits steroidogenesis in human trophoblast cells. Mol. Hum.
activin and human chorionic gonadotropin production by placental Reprod. 8, 318–325.
trophoblast cells in culture. Placenta 15, 803–818. Lytras, A., Bock, M.E., Dodd, J.G., Cattini, P.A., 1994. Detection of
Khoo, N.K., Bechberger, J.F., Shepherd, T., Bond, S.L., McCrae, K.R., placental growth hormone variant and chorionic somatomammotropin
Hamilton, G.S., Lala, P.K., 1998. SV40 Tag transformation of the ribonucleic acid expression in human trophoblastic neoplasms by
normal invasive trophoblast results in a premalignant phenotype I reverse transcriptase-polymerase chain reaction. Endocrinology 134,
Mechanisms responsible for hyperinvasiveness and resistance to anti- 2461–2467.
invasive action of TGFbeta. Int. J. Cancer 77, 429–439. Makino, Y., Hosoda, H., Shibata, K., Makino, I., Kojima, M., Kangawa, K.,
King, A., Thomas, L., Bischof, P., 2000. Cell culture models of tro- Kawarabayashi, T., 2002. Alteration of plasma ghrelin levels associated
phoblast. II. Trophoblast cell lines—a workshop report. Placenta with the blood pressure in pregnancy. Hypertension 39, 781–784.
21 (Suppl. A), S113–119. Mandl, M., Haas, J., Bischof, P., Nohammer, G., Desoye, G., 2002. Serum-
King, B.R., Smith, R., Nicholson, R.C., 2001. The regulation of human dependent effects of IGF-I and insulin on proliferation and invasion of
corticotrophin-releasing hormone gene expression in the placenta. Pep- human first trimester trophoblast cell models. Histochem. Cell Biol.
tides 22, 1941–1947. 117, 391–399.
M.H.F. Sullivan / Molecular and Cellular Endocrinology 228 (2004) 103–119 117

Manyonda, I.T., Whitley, G.S., Cartwright, J.E., 2001. Trophoblast cell Nagami, A.I., Kamitani, N., Tominaga, T., 1991. Establishement of a
lines: a response to the workshop report by King et al. Placenta 22, human trophoblastic cell line (NHT cell line) from normal early
262–263. pregnancy: formation of pseudovillus structure in vitro. In: Soma,
Margioris, A.N., Grino, M., Protos, P., Gold, P.W., Chrousos, G.P., 1988. H. (Ed.), Placenta: Basic Research for Clinical Applications. Karger,
Corticotropin-releasing hormone and oxytocin stimulate the release of Basel, pp. 68–83.
placental proopiomelanocortin peptides. J. Clin. Endocrinol. Metab. Nakanishi, T., Kohroki, J., Suzuki, S., Ishizaki, J., Hiromori, Y., Takasuga,
66, 922–926. S., Itoh, N., Watanabe, Y., Utoguchi, N., Tanaka, K., 2002. Trialkyltin
Martell, R.E., Ruddon, R.W., 1990. Patterns of human chorionic go- compounds enhance human CG secretion and aromatase activity in
nadotropin expression in untreated and 8-bromoadenosine-treated JAR human placental choriocarcinoma cells. J. Clin. Endocrinol. Metab.
choriocarcinoma cells. Endocrinology 126, 2757–2764. 87, 2830–2837.
Maruo, T., Matsuo, H., Ohtani, T., Hoshina, M., Mochizuki, M., 1986. Nestler, J.E., 1987. Modulation of aromatase and P450 cholesterol side-
Differential modulation of chorionic gonadotropin (CG) subunit mes- chain cleavage enzyme activities of human placental cytotrophoblasts
senger ribonucleic acid levels and CG secretion by progesterone in by insulin and insulin-like growth factor I. Endocrinology 121, 1845–
normal placenta and choriocarcinoma cultured in vitro. Endocrinology 1852.
119, 855–864. Nestler, J.E., 1990. Insulin-like growth factor II is a potent inhibitor of the
Maruo, T., Matsuo, H., Mochizuki, M., 1991. Thyroid hormone as a aromatase activity of human placental cytotrophoblasts. Endocrinology
biological amplifier of differentiated trophoblast function in early 127, 2064–2070.
pregnancy. Acta Endocrinol. (Copenh.) 125, 58–66. Nestler, J.E., 1993a. Regulation of the aromatase activity of human pla-
Mason, J.I., Ushijima, K., Doody, K.M., Nagai, K., Naville, D., Head, cental cytotrophoblasts by insulin, insulin-like growth factor-I, and
J.R., Milewich, L., Rainey, W.E., Ralph, M.M., 1993. Regulation of -II. J. Steroid Biochem. Mol. Biol. 44, 449–457.
expression of the 3 beta-hydroxysteroid dehydrogenases of human Nestler, J.E., 1993b. Interleukin-1 stimulates the aromatase activity of
placenta and fetal adrenal. J. Steroid Biochem. Mol. Biol. 47, 151–159. human placental cytotrophoblasts. Endocrinology 132, 566–570.
Masuhiro, K., Matsuzaki, N., Nishino, E., Taniguchi, T., Kameda, T., Ni, X., Luo, S., Minegishi, T., Peng, C., 2000. Activin A in JEG-3 cells:
Li, Y., Saji, F., Tanizawa, O., 1991. Trophoblast-derived interleukin- potential role as an autocrine regulator of steroidogenesis in humans.
1 (IL-1) stimulates the release of human chorionic gonadotropin by Biol. Reprod. 62, 1224–1230.
activating IL-6 and IL-6-receptor system in first trimester human Nickel, B.E., Cattini, P.A., 1991. Tissue-specific expression and thyroid
trophoblasts. J. Clin. Endocrinol. Metab. 72, 594–601. hormone regulation of the endogenous placental growth hormone vari-
Matsui, H., Taga, M., Kurogi, K., Hiraga, M., Suyama, K., Ohkouchi, C., ant and chorionic somatomammotropin genes in a human choriocar-
Minaguchi, H., 2000. Biological action of keratinocyte growth factor cinoma cell line. Endocrinology 128, 2353–2359.
in BeWo cells, a human choriocarcinoma cell line. J. Endocrinol. Nishino, E., Matsuzaki, N., Masuhiro, K., Kameda, T., Taniguchi, T., Tak-
Invest. 23, 19–22. agi, T., Saji, F., Tanizawa, O., 1990. Trophoblast-derived interleukin-6
Matsumoto, K., Yamamoto, T., Kurachi, H., Nishio, Y., Takeda, T., (IL-6) regulates human chorionic gonadotropin release through IL-6
Homma, H., Morishige, K., Miyake, A., Murata, Y., 1998. Human receptor on human trophoblasts. J. Clin. Endocrinol. Metab. 71, 436–
chorionic gonadotropin-alpha gene is transcriptionally activated by epi- 441.
dermal growth factor through cAMP response element in trophoblast Nishino, R., Maruo, T., Matsuo, H., Mochizuki, M., 1991. The role of
cells. J. Biol. Chem. 273, 7800–7806. signal transduction systems in the regulation of the production and
Mazlan, M., Spence-Jones, C., Chard, T., Landon, J., McLean, C., 1990. secretion of hCG (alpha, beta) by cultured human choriocarcinoma
Circulating levels of GH-releasing hormone and GH during human cells (BeWo). Nippon Naibunpi Gakkai Zasshi 67, 666–681.
pregnancy. J. Endocrinol. 125, 161–167. Nulsen, J.C., Silavin, S.L., Kao, L.C., Ringler, G.E., Kliman, H.J., Strauss
Miller, W.L., 1998. Steroid hormone biosynthesis and actions in the III, J.F., 1989. Control of the steroidogenic machinery of the human
materno-feto-placental unit. Clin. Perinatol. 25, 799–817. trophoblast by cyclic AMP. J. Reprod. Fertil. Suppl. 37, 147–153.
Moore, C.C., Hum, D.W., Miller, W.L., 1992. Identification of positive Pasquarette, M.M., Stewart, P.M., Ricketts, M.L., Imaishi, K., Mason, J.I.,
and negative placenta-specific basal elements and a cyclic adenosine 1996. Regulation of 11 beta-hydroxysteroid dehydrogenase type 2 ac-
3 ,5 -monophosphate response element in the human gene for P450scc. tivity and mRNA in human choriocarcinoma cells. J. Mol. Endocrinol.
Mol. Endocrinol. 6, 2045–2058. 16, 269–275.
Morrish, D.W., Bhardwaj, D., Dabbagh, L.K., Marusyk, H., Siy, O., Pattillo, R.A., Gey, G.O., 1968. The establishment of a cell line of human
1987. Epidermal growth factor induces differentiation and secretion hormone-synthesizing trophoblastic cells in vitro. Cancer Res. 28,
of human chorionic gonadotropin and placental lactogen in normal 1231–1236.
human placenta. J. Clin. Endocrinol. Metab. 65, 1282–1290. Pattillo, R.A., Gey, G.O., Delfs, E., Huang, W.Y., Hause, L., Garancis,
Morrish, D.W., Dakour, J., Li, H., Xiao, J., Miller, R., Sherburne, R., D.J., Knoth, M., Amatruda, J., Bertino, J., Friesen, H.G., Mattingly,
Berdan, R.C., Guilbert, L.J., 1997. In vitro cultured human term cy- R.F., 1971. The hormone-synthesizing trophoblastic cell in vitro: a
totrophoblast: a model for normal primary epithelial cells demonstrat- model for cancer research and placental hormone synthesis. Ann. N.
ing a spontaneous differentiation programme that requires EGF for Y. Acad. Sci. 172, 288–298.
extensive development of syncytium. Placenta 18, 577–585. Pedersen, A.M., Fulton, S.K., Porter, L., Francis, G.L., 1995. Tumor
Morrish, D.W., Marusyk, H., 1997. Localization of human chorionic necrosis factor-alpha affects in vitro hormone production by JEG-3
gonadoptropin and placental lactogen by immunogold labeling for choriocarcinoma cell cultures. J. Reprod. Immunol. 29, 69–80.
electron microscopy: technique and limitations. Microsc. Res. Tech. Petraglia, F., Coukos, G., Battaglia, C., Bartolotti, A., Volpe, A., Nappi,
38, 176–187. C., Segre, A., Genazzani, A.R., 1989. Plasma and amniotic fluid
Morrish, D.W., Dakour, J., Li, H., 1998. Functional regulation of human immunoreactive neuropeptide-Y level changes during pregnancy, labor,
trophoblast differentiation. J. Reprod. Immunol. 39, 179–195. and at parturition. J. Clin. Endocrinol. Metab. 69, 324–328.
Morrish, D.W., Whitley, G.S., Cartwright, J.E., Graham, C.H., Caniggia, I., Petraglia, F., Calza, L., Giardino, L., Zanni, M., Florio, P., Ferrari, A.R.,
2002. In vitro models to study trophoblast function and dysfunction— Nappi, C., Genazzani, A.R., 1993. Maternal decidua and fetal mem-
a workshop report. Placenta 23 (Suppl. A), S114–118. branes contain immunoreactive neuropeptide Y. J. Endocrinol. Invest.
Myat, T., Hitsumoto, Y., Saheki, S., Kitagawa, H., Yano, J., Matsuura, S., 16, 201–205.
1996. Attenuation of human chorionic gonadotropin release by nitric Petraglia, F., de Micheroux, A.A., Florio, P., Salvatori, M., Gallinelli,
oxide in choriocarcinoma cell lines. J. Endocrinol. 150, 243–253. A., Cela, V., Palumbo, M.A., Genazzani, A.R., 1995. Steroid-protein
118 M.H.F. Sullivan / Molecular and Cellular Endocrinology 228 (2004) 103–119

interaction in human placenta. J. Steroid Biochem. Mol. Biol. 53, growth hormone variant mRNA in the placenta. Mol. Cell. Endocrinol.
227–231. 92, R7–13.
Petraglia, F., Florio, P., Nappi, C., Genazzani, A.R., 1996. Peptide sig- Seki, H., Zosmer, A., Elder, M.G., Sullivan, M.H., 1997. The regulation of
naling in human placenta and membranes: autocrine, paracrine, and progesterone and hCG production from placental cells by interleukin-
endocrine mechanisms. Endocrinol. Rev. 17, 156–186. 1beta. Biochim. Biophys. Acta 1336, 342–348.
Petraglia, F., Santuz, M., Florio, P., Simoncini, T., Luisi, S., Plaino, Sen, K.K., Azhar, S., Menon, K.M., 1979. Receptor-mediated go-
L., Genazzani, A.R., Genazzani, A.D., Volpe, A., 1998. Paracrine nadotropin action in the ovary. Desensitization of gonadotropin binding
regulation of human placenta: control of hormonogenesis. J. Reprod. sites, activation of adenosine 3 :5 -cyclic monophosphate-dependent
Immunol. 39, 221–233. protein kinase(s), and regulation of steroidogenesis in rat ovary. J.
Potgens, A.J., Bolte, M., Huppertz, B., Kaufmann, P., Frank, H.G., 2001. Biol. Chem. 254, 5664–5671.
Human trophoblast contains an intracellular protein reactive with an Shaamash, A.H., Elsonosy, E.D., Zakhari, M.M., Radwan, S.H., El-Dien,
antibody against CD133—a novel marker for trophoblast. Placenta 22, H.M., 2001. Placental nitric oxide synthase (NOS) activity and ni-
639–645. tric oxide (NO) production in normal pregnancy, pre-eclampsia and
Qu, J., Thomas, K., 1998. Advance in the study of inhibin, activin and eclampsia. Int. J. Gynaecol. Obstet. 72, 127–133.
follistatin production in pregnant women. Eur. J. Obstet. Gynecol. Shi, C.Z., Zhang, Z.Y., Zhuang, L.Z., 1991. Study on reproductive en-
Reprod. Biol. 81, 141–148. docrinology of human placenta (III)—hormonal regulation of proges-
Reis, F.M., Petraglia, F., 2001. The placenta as a neuroendocrine organ. terone production by trophoblast tissue of first trimester. Sci. Chin. B
Front. Horm. Res. 27, 216–228. 34, 1098–1104.
Reis, F.M., Florio, P., Cobellis, L., Luisi, S., Severi, F.M., Bocchi, C., Shi, C.Z., Zhuang, L.Z., 1993a. Norepinephrine regulates human chorionic
Picciolini, E., Centini, G., Petraglia, F., 2001. Human placenta as a gonadotrophin production by first trimester trophoblast tissue in vitro.
source of neuroendocrine factors. Biol. Neonate. 79, 150–156. Placenta 14, 683–693.
Ren, S.G., Braunstein, G.D., 1991. Insulin stimulates synthesis and re- Shi, C.Z., Zhuang, L.Z., 1993b. Stimulatory effect of norepinephrine on
lease of human chorionic gonadotropin by choriocarcinoma cell lines. progesterone production by human first trimester placenta explants in
Endocrinology 128, 1623–1629. vitro. Life Sci. 52, 1657–1665.
Shih, I., Wang, T., Wu, T., Kurman, R.J., Gearhart, J.D., 1998. Expression
Ringler, G.E., Kao, L.C., Miller, W.L., Strauss III, J.F., 1989. Effects
of Mel-CAM in implantation site intermediate trophoblastic cell line,
of 8-bromo-cAMP on expression of endocrine functions by cultured
IST-1, limits its migration on uterine smooth muscle cells. J. Cell Sci.
human trophoblast cells. Regulation of specific mRNAs. Mol. Cell.
111 (17), 2655–2664.
Endocrinol. 61, 13–21.
Shimada, A., Nakano, H., Takahashi, T., Imai, K., Hashizume, K., 2001.
Risk, J.M., Johnson, P.M., 1990. Northern blot analysis of HLA-G ex-
Isolation and characterization of a bovine blastocyst-derived tro-
pression by BeWo human choriocarcinoma cells. J. Reprod. Immunol.
phoblastic cell line, BT-1: development of a culture system in the
18, 199–203.
absence of feeder cell. Placenta 22, 652–662.
Ritvos, O., Jalkanen, J., Pekonen, F., Stenman, U.H., Ranta, T., 1988. Epi-
Shimonovitz, S., Hurwitz, A., Hochner-Celnikier, D., Dushnik, M., An-
dermal growth factor but not insulin-like growth factor-I potentiates
teby, E., Yagel, S., 1998. Expression of gelatinase B by trophoblast
adenosine 3 ,5 -monophosphate-mediated chorionic gonadotropin se-
cells: down-regulation by progesterone. Am. J. Obstet. Gynecol. 178,
cretion by cultured human choriocarcinoma cells. Endocrinology 123,
457–461.
859–865.
Shiverick, K.T., King, A., Frank, H., Whitley, G.S., Cartwright, J.E.,
Ritvos, O., Eramaa, M., 1991. Adenosine 3 ,5 -monophosphate and phor-
Schneider, H., 2001. Cell culture models of human trophoblast. II.
bol ester induce transforming growth factor-beta 1 messenger ribonu-
Trophoblast cell lines—a workshop report. Placenta 22 (Suppl. A),
cleic acid levels in choriocarcinoma cells. Endocrinology 129, 2240–
S104–106.
2245.
Sibley, C.P., Hochberg, A., Boime, I., 1991. Bromo-adenosine stimulates
Rong-Hao, L., Luo, S., Zhuang, L.Z., 1996. Establishment and character- choriogonadotropin production in JAr and cytotrophoblast cells: evi-
ization of a cytotrophoblast cell line from normal placenta of human dence for effects on two stages of differentiation. Mol. Endocrinol. 5,
origin. Hum. Reprod. 11, 1328–1333. 582–586.
Saito, S., Ibaraki, T., Enomoto, M., Ichijo, M., Motoyoshi, K., 1994. Siler-Khodr, T.M., Kang, I.S., Koong, M.K., Grayson, M., 1997. The
Macrophage colony-stimulating factor induces the growth and differ- effect of dexamethasone on CRH and prostanoid production from
entiation of normal pregnancy human cytotrophoblast cells and hyda- human term placenta. Prostaglandins 54, 639–653.
tidiform moles but does not induce the growth and differentiation of Sorem, K.A., Smikle, C.B., Spencer, D.K., Yoder, B.A., Graveson, M.A.,
choriocarcinoma cells. Jpn. J. Cancer Res. 85, 245–252. Siler-Khodr, T.M., 1996. Circulating maternal corticotropin-releasing
Salido, E.C., Yen, P.H., Barajas, L., Shapiro, L.J., 1990. Steroid sulfa- hormone and gonadotropin-releasing hormone in normal and abnormal
tase expression in human placenta: immunocytochemistry and in situ pregnancies. Am. J. Obstet. Gynecol. 175, 912–916.
hybridization study. J. Clin. Endocrinol. Metab. 70, 1564–1567. Stephanou, A., Handwerger, S., 1994. Interleukin-6 stimulates placental
Sanyal, M., Das, C., 1997. Collagenase-IV in human trophoblast invasion lactogen expression by human trophoblast cells. Endocrinology 135,
and differentiation. Indian J. Biochem. Biophys. 34, 220–225. 719–723.
Sarkar, S., Tsai, S.W., Nguyen, T.T., Plevyak, M., Padbury, J.F., Rubin, Stephanou, A., Handwerger, S., 1995. Retinoic acid and thyroid hormone
L.P., 2001. Inhibition of placental 11 beta-hydroxysteroid dehydroge- regulate placental lactogen expression in human trophoblast cells.
nase type 2 by catecholamines via alpha-adrenergic signaling. Am. J. Endocrinology 136, 933–938.
Physiol. Regul. Integr. Comp. Physiol. 281, R1966–1974. Stewart, P.M., Rogerson, F.M., Mason, J.I., 1995. Type 2 11 beta-
Scatena, C.D., Adler, S., 1996. Trans-acting factors dictate the species- hydroxysteroid dehydrogenase messenger ribonucleic acid and activ-
specific placental expression of corticotropin-releasing factor genes in ity in human placenta and fetal membranes: its relationship to birth
choriocarcinoma cell lines. Endocrinology 137, 3000–3008. weight and putative role in fetal adrenal steroidogenesis. J. Clin. En-
Schneider-Kolsky, M.E., Manuelpillai, U., Waldron, K., Dole, A., Wal- docrinol. Metab. 80, 885–890.
lace, E.M., 2002. The distribution of activin and activin receptors in Strauss III, J.F., Kido, S., Sayegh, R., Sakuragi, N., Gafvels, M.E.,
gestational tissues across human pregnancy and during labour. Pla- 1992. The cAMP signalling system and human trophoblast function.
centa 23, 294–302. Placenta 13, 389–403.
Scippo, M.L., Frankenne, F., Hooghe-Peters, E.L., Igout, A., Velkeniers, Strauss III, J.F., Christenson, L.K., Devoto, L., Martinez, F., 2000.
B., Hennen, G., 1993. Syncytiotrophoblastic localization of the human Providing progesterone for pregnancy: control of cholesterol flux
M.H.F. Sullivan / Molecular and Cellular Endocrinology 228 (2004) 103–119 119

to the side-chain cleavage system. J. Reprod. Fertil. Suppl. 55, 3– Yagel, S., Lala, P.K., Powell, W.A., Casper, R.F., 1989. Interleukin-1
12. stimulates human chorionic gonadotropin secretion by first trimester
Strohmer, H., Kiss, H., Mosl, B., Egarter, C., Husslein, P., Knofler, M., human trophoblast. J. Clin. Endocrinol. Metab. 68, 992–995.
1997. Hypoxia downregulates continuous and interleukin-1-induced Yamamoto, T., Roby, K.F., Kwok, S.C., Soares, M.J., 1994. Transcriptional
expression of human chorionic gonadotropin in choriocarcinoma cells. activation of cytochrome P450 side chain cleavage enzyme expression
Placenta 18, 597–604. during trophoblast cell differentiation. J. Biol. Chem. 269, 6517–6523.
Sun, K., Yang, K., Challis, J.R., 1997. Differential regulation of 11 Yamamoto, T., Chapman, B.M., Clemens, J.W., Richards, J.S., Soares,
beta-hydroxysteroid dehydrogenase type 1 and 2 by nitric oxide in M.J., 1995. Analysis of cytochrome P-450 side-chain cleavage gene
cultured human placental trophoblast and chorionic cell preparation. promoter activation during trophoblast cell differentiation. Mol. Cell.
Endocrinology 138, 4912–4920. Endocrinol. 113, 183–194.
Sun, K., Yang, K., Challis, J.R., 1998. Regulation of 11 beta- Yamamoto, T., Matsumoto, K., Kurachi, H., Okamoto, Y., Nishio, Y.,
hydroxysteroid dehydrogenase type 2 by progesterone, estrogen, and Sakata, M., Tasaka, K., Murata, Y., 2001. Progesterone inhibits tran-
the cyclic adenosine 5 -monophosphate pathway in cultured human scriptional activation of human chorionic gonadotropin-alpha gene
placental and chorionic trophoblasts. Biol. Reprod. 58, 1379–1384. through protein kinase A pathway in trophoblast cells. Mol. Cell.
Sun, K., He, P., Yang, K., 2002. Intracrine induction of 11 beta- Endocrinol. 182, 215–224.
hydroxysteroid dehydrogenase type 1 expression by glucocorticoid po- Yanushpolsky, E.H., Ozturk, M., Polgar, K., Berkowitz, R.S., Hill, J.A.,
tentiates prostaglandin production in the human chorionic trophoblast. 1993. The effects of cytokines on human chorionic gonadotropin
Biol. Reprod. 67, 1450–1455. (hCG) production by a trophoblast cell line. J. Reprod. Immunol. 25,
Tarrade, A., Lai Kuen, R., Malassine, A., Tricottet, V., Blain, P., Vidaud, 235–247.
M., Evain-Brion, D., 2001. Characterization of human villous and Yui, J., Garcia-Lloret, M., Brown, A.J., Berdan, R.C., Morrish, D.W.,
extravillous trophoblasts isolated from first trimester placenta. Lab. Wegmann, T.G., Guilbert, L.J., 1994. Functional, long-term cultures
Invest. 81, 1199–1211. of human term trophoblasts purified by column-elimination of CD9
Taylor, C.M., Stevens, H., Anthony, F.W., Wheeler, T., 1997. Influence expressing cells. Placenta 15, 231–246.
of hypoxia on vascular endothelial growth factor and chorionic go- Zdravkovic, M., Aboagye-Mathiesen, G., Guimond, M.J., Hager, H., Ebbe-
nadotrophin production in the trophoblast-derived cell lines: JEG. JAr sen, P., Lala, P.K., 1999. Susceptibility of MHC class I expressing
and BeWo. Placenta 18, 451–458. extravillous trophoblast cell lines to killing by natural killer cells.
Taylor, R.N., Newman, E.D., Chen, S.A., 1991. Forskolin and methotrex- Placenta 20, 431–440.
ate induce an intermediate trophoblast phenotype in cultured human Zeitler, P., Markoff, E., Handwerger, S., 1983. Characterization of the
choriocarcinoma cells. Am. J. Obstet. Gynecol. 164, 204–210. synthesis and release of human placental lactogen and human chorionic
Tonkowicz, P.A., Poisner, A.M., 1985. Evidence for a role for adenosine gonadotropin by an enriched population of dispersed placental cells.
3 ,5 -monophosphate in progesterone secretion by human chorion. J. Clin. Endocrinol. Metab. 57, 812–818.
Endocrinology 116, 646–650. Zeitler, P., Handwerger, S., 1985. Arachidonic acid stimulates phos-
Tsakonas, D.P., Nicolaides, K.H., Tsakona, C.P., Worman, C.P., Gold- phoinositide hydrolysis and human placental lactogen release in
stone, A.H., 1995. Changes in maternal plasma macrophage-colony an enriched fraction of placental cells. Mol. Pharmacol. 28, 549–
stimulating factor levels during normal pregnancy. Clin. Lab. Haema- 554.
tol. 17, 57–59. Zeitler, P., Wu, Y.Q., Handwerger, S., 1991. Melittin stimulates phospho-
Tuckey, R.C., Kostadinovic, Z., Cameron, K.J., 1994. Cytochrome P- inositide hydrolysis and placental lactogen release: arachidonic acid
450scc activity and substrate supply in human placental trophoblasts. as a link between phospholipase A2 and phospholipase C signal-
Mol. Cell. Endocrinol. 105, 123–129. transduction pathways. Life Sci. 48, 2089–2095.
Ugele, B., Simon, S., 1999. Uptake of dehydroepiandrosterone-3-sulfate Zhang, L., Connor, E.E., Chegini, N., Shiverick, K.T., 1995. Modulation by
by isolated trophoblasts from human term placenta, JEG-3, BeWo, benzo[a]pyrene of epidermal growth factor receptors, cell proliferation,
Jar, BHK cells, and BHK cells transfected with human sterylsulfatase- and secretion of human chorionic gonadotropin in human placental
cDNA. J. Steroid Biochem. Mol. Biol. 71, 203–211. cell lines. Biochem. Pharmacol. 50, 1171–1180.
Wang, L., Zhang, W., Zhao, Y., 1999. The study of maternal and fetal Zhou, F., Yuen, B.H., Leung, P.C., 1987. Effects of adenosine 3 ,5 -cyclic
plasma catecholamines levels during pregnancy and delivery. J. Perinat. monophosphate forskolin and cholera toxin on hormone production in
Med. 27, 195–198. human term placental cells. Am. J. Obstet. Gynecol. 156, 420–424.
Wunsch, D.M., Anderson, L.D., Pepe, G.J., Albrecht, E.D., 1986. Regu- Zosmer, A., Elder, M.G., Sullivan, M.H., 1997. Second messengers and
lation of progesterone formation by human placental cells in culture. the control of progesterone production from first trimester trophoblast.
Endocrinology 119, 998–1003. J. Steroid Biochem. Mol. Biol. 62, 201–205.

You might also like