2015 - Physiology Production and Action of Progesterone

You might also like

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

A C TA Obstetricia et Gynecologica

AOGS RE V I EW AR TIC LE

Physiology, production and action of progesterone


STEFANIA TARABORRELLI, MD
Reproductive Medicine Unit, GynePro Medical Group, Bologna, Italy

Key words Abstract


Progesterone synthesis, progesterone effects,
window of implantation, genomic and non- Introduction. The aim of this article is to review the physiology of proges-
genomic action terone and focus on its physiological actions on tissues such as endome-
trium, uterus, mammary gland, cardiovascular system, central nervous
Correspondence system and bones. In the last decades, the interest of researchers has
Stefania Taraborrelli, GynePro Medical Group,
focused on the role of progesterone in genomic and non-genomic receptor
Via T. Cremona 8, 40137 Bologna, Italy.
mechanisms. Materials and Methods. We searched PubMed up to December
E-mail: s.taraborrelli@gynepro.it
2014 for publications on progesterone/steroidogenesis. Results and Conclu-
Conflict of interest sions. A better understanding of the biological genomic and non-genomic
The author has stated explicitly that there are receptor mechanisms could enable us in the near future to obtain a more
no conflicts of interest in connection with this comprehensive knowledge of the safety and efficacy of this agent during
article. hormone replacement therapy (natural progesterone), in vitro fertilization
(water-soluble subcutaneous progesterone), in traumatic brain injury, Alzhei-
Please cite this article as: Taraborrelli S.
mer’s disease and diabetic neuropathy, even though further clinical studies
Physiology, production and action of
progesterone. Acta Obstet Gynecol Scand are needed to prove its usefulness.
2015; 94: 8–16.
Abbreviations: DOC, desoxycorticosterone; FSH, follicle-stimulating hormone;
Received: 17 March 2015 HDL, high-density lipoprotein; HRT, hormonal replacement therapy; LDL,
Accepted: 3 September 2015
low-density lipoprotein; LH, luteinizing hormone; nPR, nuclear progesterone
DOI: 10.1111/aogs.12771
receptor; PGRMC, progesterone receptor membrane component; PR,
progesterone receptor; SERBP1, serpinel mRNA binding protein I.

essary to achieve a sufficient biological effect. The


development of galenic formulations allowed proges-
Introduction
terone to be used in hormonal therapies (1). In the
Progesterone allows the endometrial transition from a 1950s, Birch obtained a compound with a progestin
proliferative to the secretory stage, facilitates blastocyst activity that was four to eight times higher than pro-
nesting and is essential to the maintenance of pregnancy. gesterone, replacing the methyl group in position 19
These characteristics explain the ethymology of the hor- with a hydrogen atom (19 nor-progesterone); the for-
mone0 s name, which comes from the Latin pro gesta- mulation of numerous other progestin compounds fol-
tionem. lowed this (2).
Progesterone also plays an important role in several
tissues not belonging to the reproductive system, such
as the mammary gland in preparation for breastfeeding,
the cardiovascular system, central nervous system and
bones. Allen originally described the molecular formula Key Message
of progesterone in 1933; however, only in the mid- A better understanding of the biological actions of
1940s did Russell Marker begin to synthesize proges- progesterone could enable safe and effective use of
terone from diosgenin, extracted from the Japanese this hormone in different scientific and medical
plant Dioscorea tokoro, and later from Dioscorea mexi- fields.
cana. At first, high concentrations of extract were nec-

8 ª 2015 Nordic Federation of Societies of Obstetrics and Gynecology, Acta Obstetricia et Gynecologica Scandinavica 94 (2015) 8–16
16000412, 2015, S161, Downloaded from https://obgyn.onlinelibrary.wiley.com/doi/10.1111/aogs.12771 by Nat Prov Indonesia, Wiley Online Library on [25/07/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Stefania Taraborrelli Physiology of progesterone

Methods • the D4-ketosteroid pathway, which leads to the synthe-


sis of 17 a-hydroxyprogesterone and androstenedione,
We searched PubMed up to December 2014 for publica-
and is characteristic of corpus luteum granulosa cells
tions on progesterone/steroidogenesis, with regard to the
(Figure 1).
ovary, endometrium, and non-genomic and genomic
receptor information. Whereas progesterone produced from the gonads is car-
ried mostly in the blood to exert its biological function,
progesterone of adrenal origin is largely converted into
Progesterone from cycle to glucocorticoids and androgens (7). Progesterone circulates
pregnancy in the bloodstream bound to cortisol-binding globulin
Steroidogenesis (about 10%) and serum albumin. Progesterone has a rela-
tively short half-life in the body of only five minutes. The
Steroids are ancestral molecules developed about 2 billion metabolites mainly produced in the liver are sulfates and
years ago as primitive bioregulators between primordial glucuronides of reduced derivates and are excreted in the
cells (3). Steroid hormones are characterized by a com- urine. Circulating progesterone is converted to the potent
mon basic structure of cyclopentane-perhydro-phenan- mineralocorticoid desoxycorticosterone (DOC) by renal
trene, a polycyclic complex of 17 carbon atoms forming a 21-hydroxylation. During the luteal phase, during preg-
four-ring system. According to the number of carbon nancy and during exogenous progesterone administration,
atoms, sex steroids are divided into three groups: most circulating DOC arises via this pathway and may
• progestins, characterized by 21-carbon atoms; constitute an unwanted side-effect (7).
• androgens, characterized by 19-carbon atoms;
• estrogens, characterized by 18-carbon atoms. Pharmacokinetics of progesterone
The biosynthetic pathway of steroid hormones is the Although the route of transdermal administration of pro-
same regardless of the steroid-generating organ (ovary, gesterone would offer good patient compliance, it cannot
testis, adrenal cortex, brain and placenta), but the type provide an adequate plasma level of circulating proges-
and the amount of synthesized and secreted steroids terone, and, in addition, the sublingual administration
depends on the expression of enzymes specific to each of route is inconvenient because more daily doses are
these organs. required.
The female gonad, despite being a complete steroido- Orally administered progesterone is absorbed through
genic gland, differs from the other organs of this type in the bowel and reaches the liver via the portal vein, where
enzymatic provision and then in hormones secreted; the it is rapidly converted to metabolites: this is why high
ovaries do not contain 21 a-hydroxylase or 11 a-hydroxy- oral progesterone levels are needed to achieve adequate
lase, so they are not able to produce glucocorticoids or plasma levels.
mineralcorticoids (4). In contrast, the vaginal administration route, even if it
Cholesterol is the common precursor of all steroid hor- does not allow high serum levels to be achieved, has a
mones. Its synthesis starts with the interconversion of two preferential distribution to the uterus and then into the
molecules of acetyl-CoA and proceeds through the forma- endometrium. Although intramuscular administration has
tion of the two intermediate products squalene and lanes- poor patient compliance, it remains the administration
terol (5). Cholesterol conveyed in plasma as low-density route that ensures the achievement of adequate plasma
lipoprotein (LDL) cholesterol (6) interacts with mem- levels that can be monitored by serum level measure-
brane receptors and is internalized in vesicles and then ments. In future, a subcutaneous administration route,
fused with lysosomes. The lysosomal hydrolases allow the which is clearly easier for the patient and thus ensures
intracellular release of free form cholesterol, which is better compliance, may become available.
transported into the mitochondria to be subsequently
converted to pregnenolone. Pregnenolone released from
mitochondria through the action of the cytochrome P450 Role of progesterone in different stages of the
can follow two metabolic pathways: ovarian cycle

• the D5-hydroxy steroid pathway, which leads to the To understand the crucial effect of progesterone on men-
strual cycle physiology, it is necessary to refer briefly to
synthesis of 17 a-hydroxypregnenolone, dehy-
droepiandrosterone (DHEA) and androstenediol, and is the well-known “two-cell-two-gonadotrophin theory”
the main metabolic pathway in adrenal glands and not described for the first time at the end of the 1960s by
luteinized follicle, Ryan and Petro (8).

ª 2015 Nordic Federation of Societies of Obstetrics and Gynecology, Acta Obstetricia et Gynecologica Scandinavica 94 (2015) 8–16 9
16000412, 2015, S161, Downloaded from https://obgyn.onlinelibrary.wiley.com/doi/10.1111/aogs.12771 by Nat Prov Indonesia, Wiley Online Library on [25/07/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Physiology of progesterone Stefania Taraborrelli

Figure 1. Steroidogenesis, an overview.

The onset of estrogen synthesis in antral and pre- During ovulation, which occurs about 34–36 h after
antral follicles is enabled by the close functional relation the LH surge, the secondary oocyte in metaphase II leaves
between granulosa and theca cells. In fact, through the the dominant follicle and passes into the Fallopian tube
action of luteinizing hormone (LH), theca cells promote lumen, where it can be fertilized. The dominant follicle
the conversion of serum cholesterol to androstenedione, becomes the corpus luteum, a dynamic, atypical and tem-
which is then converted to estrogens by the action of porary endocrine structure. Dominant follicle luteiniza-
follicle-stimulating hormone and by the activity of the tion by LH occurs through the action of angiogenic
enzymatic aromatase system (9). The preovulatory folli- factors contained in follicular fluid (11), which allows the
cle is able to synthesize estrogens as well as progesterone proliferation of blood capillaries and fibroblasts to the
which act directly on granulosa cells by promoting fol- basal lamina of granulosa cells. This mechanism appears
licular growth and by inhibiting apoptotic genes, to be essential to allow LDL cholesterol to reach the lutei-
through interaction with a membrane receptor (Proges- nized cells and then be converted to progesterone
terone Receptor Membrane Component-1, PGRMC1) through the action of cytochrome P450 and a-hydroxy
(10). dehydrogenase (12). In a normal menstrual cycle, proges-

10 ª 2015 Nordic Federation of Societies of Obstetrics and Gynecology, Acta Obstetricia et Gynecologica Scandinavica 94 (2015) 8–16
16000412, 2015, S161, Downloaded from https://obgyn.onlinelibrary.wiley.com/doi/10.1111/aogs.12771 by Nat Prov Indonesia, Wiley Online Library on [25/07/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Stefania Taraborrelli Physiology of progesterone

terone concentrations during the follicular phase are defined as the time window in which the blastocyst
lower than 1 ng/mL, but in the days following ovulation adheres to the endometrial cells and then proceeds to
they reach values between 10 and 35 ng/mL. invade the stroma. At the time of conception and in early
In the luteal phase, progesterone secretion is episodic pregnancy, progesterone concentrations do not show sub-
and its variations are closely correlated with the LH pul- stantial changes, but they tend to increase later, reaching
satile release. However, in the early luteal phase the pat- values of 100–300 ng/mL in term pregnancy.
tern of progesterone secretion is stable throughout the Progesterone is almost entirely produced by the corpus
day and is not correlated to LH pulses; conversely, in the luteum until the ninth week of pregnancy (17); thereafter
mid- and late luteal phase the frequency of LH pulses the trophoblasts increase progesterone production, thus
decreases while the amplitude of LH pulse increases. becoming the largest source of this hormone after the
Thus, progesterone levels peak during the mid-luteal 12th week of gestation (18). Corpus luteum deficiency
phase and decline in the late luteal phase (13). In the seems to occur in 35% of recurrent miscarriage cases
absence of conception, the corpus luteum involves rapidly (18). Progesterone is primarily synthetized from maternal
within 9–11 days after ovulation. The mechanism of cor- LDL cholesterol by the placenta through a complete
pus luteum degeneration has not been exactly clarified; enzyme system and only a small fraction comes from fetal
reduced sensitivity to LH and the actions of prostaglan- steroidogenesis. Progesterone affects tubal motility by
dins and cytokines seem to play an important role (13). interacting on specific receptors and acts on endometrial
maturation and on uterine vascularization in the pre-im-
plantation phase (18). Together with human chorionic
Endometrial effects of progesterone during the
gonadotrophin and decidual cortisol it inhibits the T
menstrual cycle
lymphocyte-mediated tissue reaction (18). Progesterone
Progesterone plays a very important role in the transition seems to induce tocolytic and immunosuppressive effects
of the endometrium from the proliferative to the secre- in the areas of major contact between maternal and fetal
tory phase. The expression of endometrial receptors for compartments (18).
estrogens and progesterone changes during the various
phases of the physiological menstrual cycle. During the
Mechanisms of genomic and non-
proliferative phase, there is a predominance of estrogen
genomic action of progesterone
receptors localized in stromal and myometrial epithelial
cells, but during the ovulatory phase this concentration In the last decade, the interest of researchers has focused
falls rapidly due to the suppressive action of progesterone. on the role of progesterone in genomic (nuclear) and
Conversely, progesterone receptors (PRs), mainly local- non-genomic (extranuclear) receptor mechanisms, which
ized in endometrial epithelial cells, increase exponentially cooperate to produce direct effects on cells and tissues.
in the ovulatory phase, as a result of the estradiol action,
and then drastically decrease in the late luteal phase (14).
Genomic receptor mechanism (nuclear)
During the secretory phase, the endometrium is subject
to the combined action of estrogens and progesterone. Progesterone, being a lipophilic molecule, readily crosses
Its0 growth stops with a decline in cellular mitotic activ- cell membranes by diffusion and interacts at the nuclear
ity, mainly due to the action of progesterone. To convert level with the specific PRs, PR-B and PR-A, thus activat-
estradiol to estrone sulfate two enzyme systems are neces- ing about 300 distinct co-regulators that act on riboso-
sary, i.e. 17 a-OH-SD (estradiol dehydrogenase) and sul- mal RNA (19) and result in the production of
fotransferase (15). Moreover, progesterone administration corresponding proteins (20). Nuclear progesterone recep-
during hormonal replacement therapy (HRT) would tors (nPR) take minutes or hours to activate ribosomal
appear to confirm the protective effect of this hormone transcription and are the main regulators of female
on the endometrium of peri- and postmenopausal reproduction (21).
women, as indicated in the Postmenopausal Estrogen/ Genomic receptors PR-B and PR-A (Figure 2) share
Progestin Interventions Study (PEPI trial) (16). the central portion of the DNA binding domain and
the carboxy-terminal end of the ligand binding domain
but they differ in amino acid sequence, PR-A having
Effects of progesterone from the window of
164 amino acids fewer than PR-B (21,22). Under physi-
implantation to pregnancy
ological conditions in humans, cells expressing PR-B
The embryo enters the uterus at the morula stage about and PR-A are equally represented. However, a third iso-
two to three days after fertilization and implantation form of nPR, PR-C, is abundantly found in myometrial
begins five to six days later. Endometrial receptivity is tissue (23).

ª 2015 Nordic Federation of Societies of Obstetrics and Gynecology, Acta Obstetricia et Gynecologica Scandinavica 94 (2015) 8–16 11
16000412, 2015, S161, Downloaded from https://obgyn.onlinelibrary.wiley.com/doi/10.1111/aogs.12771 by Nat Prov Indonesia, Wiley Online Library on [25/07/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Physiology of progesterone Stefania Taraborrelli

Non-genomic receptor mechanism (extranuclear)


In 1942, Hans Selye reported that intraperitoneal injection
of progesterone in rats induced a prompt anesthetic effect,
showing for the first time the rapid non-genomic action of
progesterone at tissue level (24). Historically, two models
were studied in a comprehensive way to evaluate proges-
terone non-genomic effects: sperm acrosome reaction and
oocyte maturation induction in animal models such as
Xenopus laevis (25). Over the years, an interesting observa-
tion was made: the rapid effect of the non-genomic pro-
gesterone-R interaction in various tissues activates a wide
variety of secondary messengers (Table 1).
The progesterone non-genomic effects present the fol-
lowing characteristics:
Figure 2. Progesterone receptor (PR) A, B and C structures. (From
Gellersen B, Fernandes MS, Brosens JJ. Non-genomic progesterone • they are too rapid to be compared to the transcrip-
actions in female reproduction. Hum Reprod Update. 2009;15;119– tional effects of the other genomic effects (21);
38. © 2009 European Society of Human Reproduction and • they are found in specific cellular compartments that
Embryology. Reproduced with permission by Oxford University Press lacks cell nuclei such as erythrocytes or platelets (21);
on behalf of the European Society of Human Reproduction and
Embryology).
• they are not suppressed by inhibitors of steroid nuclear
receptors (21).

Table 1. Progesterone effects on tissues (from Gellersen B, Fernandes MS, Brosens JJ. Non-genomic progesterone actions in female
reproduction. Hum Reprod Update. 2009;15;119–38. © 2009 European Society of Human Reproduction and Embryology. Reproduced with
permission by Oxford University Press on behalf of the European Society of Human Reproduction and Embryology).*

Physiological action Cell/tissue/organism Signaling pathway References


2+ +
Acrosome reaction/ Human spermatozoa Ca influx, Cl efflux, cAMP increase Luconi (2004), Blackmore (1991),
capacitation Kirkman-Brown (2000)
Oocyte maturation Amphibian and fish oocytes G-protein activation and cAMP Zhu et al. (2003b), Thomas et al.
decrease, ERK1/2 activation, PI3 (2002), Maller (2001), Bagowski
kinase activation (2001)
Immunoregulatory function Human T-lymphocytes G-protein activation, K+ channel Dosiou (2008), Ehring (1998)
inhibition
Platelet aggregation Human platelets Ca2+ influx Bar (2000), Blackmore (1999, 2008)
Anti-apoptotic effects Rat granulosa cells MAPK kinase (MEK) inhibition, Ca2+ Peluso (2001), Peluso et al. (2004)
homeostasis, Protein kinase G
activation
Muscle contraction Human intestinal smooth Ca2+ currents reduction Bielefeldt (1996)
muscle cells
Vasoreactivity Rat vascular smooth muscle cells Ca2+ influx regulation Barbagallo (2001)
Steroidogenesis and LH Rodent Leydig cells Na+ influx Rossato (1999), El-Hefnawy and
action Huhtaniemi (1998)
Lordosis Female mice Frye (2006)
Transepithelial resistance Human fetal membranes Not assessed Verikouki (2008)
Actin cytoskeleton- Human umbilical vein G-protein activation, PI3 kinase and Fu (2008a, b)
remodeling/cell endothelial cells, human RhoA/ROCK-2 cascade activation
movement breast cancer cells
Neuroprotection Mouse cerebral cortex, rat PI3 kinase activation, ERK1/2 activation, Kaur (2007), Nilsen and Brinton
hippocampal neurons Ca2+ influx inhibition (2003), Cai (2008)
Retinal neuronal activity Mouse rod bipolar cells PI3 kinase activation Koulen (2008)

*
All bibliographic references included in the Table are listed in the References of the article (44–67).

12 ª 2015 Nordic Federation of Societies of Obstetrics and Gynecology, Acta Obstetricia et Gynecologica Scandinavica 94 (2015) 8–16
16000412, 2015, S161, Downloaded from https://obgyn.onlinelibrary.wiley.com/doi/10.1111/aogs.12771 by Nat Prov Indonesia, Wiley Online Library on [25/07/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Stefania Taraborrelli Physiology of progesterone

0.25–0.5 mg/day results in a marked improvement in bone


mineral density in postmenopausal women (30); similarly,
continuous transdermal HRT with 17 a-estradiol 0.05 mg/
day and norethisterone acetate 0.25 mg/day for 14–28 days
shows a clear reduction of bone turnover. Conversely,
micronized progesterone alone in both depot and transder-
mal formulations (31) does not seem to have a positive
biological effect on bone metabolism. Recent studies have
shown that the inhibition of nPRs in rats during the period
of rapid bone growth (1–3 months) increases osteogenesis.
These results seem to suggest a new approach in the man-
Figure 3. Progesterone receptor membrane component 1 (PGRMC1)
agement of osteoporosis and further studies may support
structure. SERBP1: serpinel mRNA binding protein I. (From Gellersen
B, Fernandes MS, Brosens JJ. Non-genomic progesterone actions in its therapeutic benefit in osteoporosis (32).
female reproduction. Hum Reprod Update. 2009;15;119–38. © 2009
European Society of Human Reproduction and Embryology.
Progesterone and the central nervous system
Reproduced with permission by Oxford University Press on behalf of
the European Society of Human Reproduction and Embryology). Progesterone acts on the hypothalamic-pituitary-adreno-
cortical axis by modulating LH secretion, thereby establish-
The progesterone non-genomic receptor localized in ing a feedback mechanism on ovarian steroidogenesis (33).
the cell membrane is called PGRMC1 and has predomi- Progesterone seems to have an inhibitory effect on sexual
nantly been studied in the ovary (Figure 3). The interac- desire (34) in women and it is metabolized in 5a-dihydro-
tion between PGRMC1 and serpinel mRNA binding progesterone and allopregnanolone, the latter compound
protein I (SERBP1) determines the anti-apoptotic effect exerting neuroprotective and restorative effects on trau-
of progesterone on granulosa cells (26). It also seems that matic and ischemic brain injuries (34), where it appears to
PGRMC1, along with SERBP1, is involved in ovarian car- reduce edema and restore the function of the blood barrier.
cinogenesis and its level of expression is potentially asso- Therefore, progesterone must be regarded as a ‘neuros-
ciated with tumor invasion and metastasis (27). Less teroid’ and its metabolite allopregnanolone could also be
information is available on PGRMC2, although its expres- active on glial cells by promoting myelin production and
sion, as well as that of PGRMC1, appears under the con- slowing the progression of Alzheimer’s disease (35). More-
trol of gonadal steroids (27). over, progesterone seems to affect the cognitive and behav-
ioral spheres: increased levels of this hormone postpartum
may explain the reduction of aggressive behavior in lactating
Progesterone and other target tissues
mice (36). Therefore, progesterone seems to interact with
In addition to its clear effects on reproductive sphere, pro- non-genomic gamma-aminobutyric acid (GABA) receptors
gesterone has multiple functions on various organs and it and, in the future, this agent could be regarded as a thera-
therefore deserves to be considered a “neurosteroid”. peutic alternative in psychiatric aggressive behavior, depres-
sion and anxiety (36). We report here just a mention on the
progesterone role on premenstrual syndrome and premen-
Progesterone and osteoporosis
strual dysphoric disorder (a severe form of premenstrual
Progesterone and synthetic progestins have no or only syndrome).One theory is focused on a “corpus luteum
minor effects on bone. Its derivatives appear to have a lim- insufficiency” which creates a deficiency of progesterone
ited effect on bone density. Progestin receptors have been during this menstrual cycle phase. It has been demonstrated
identified on osteoclasts and osteoblasts (28) and it has a negative effect of low progesterone level on the premen-
been hypothesized that progesterone has a trophic action strual mood symptoms such as aggressive behavior and fati-
on the bone similar to estrogens, inhibiting bone resorp- gue in healthy reproductive age women (37) This might
tion through a direct stimulatory effect on calcitonin secre- explain the improvement of premenstrual syndrome and
tion (29). Progesterone, however, has only minimal effects premenstrual dysphoric disorder symptoms by using pro-
on bone mineral density compared to estradiol, whereas gesterone administration (38).
medroxyprogesterone acetate (MPA) and estradiol induce
a significant decrease of osteoclast activity. These factors
Progesterone and metabolic effects
should be taken into account when choosing progestin for
hormone replacement therapy. The oral combination of 17 Progesterone increases insulin basal levels and promotes
a-estradiol 1 mg/day and low dose norethisterone acetate insulin release after carbohydrate intake (4). The carpal

ª 2015 Nordic Federation of Societies of Obstetrics and Gynecology, Acta Obstetricia et Gynecologica Scandinavica 94 (2015) 8–16 13
16000412, 2015, S161, Downloaded from https://obgyn.onlinelibrary.wiley.com/doi/10.1111/aogs.12771 by Nat Prov Indonesia, Wiley Online Library on [25/07/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Physiology of progesterone Stefania Taraborrelli

tunnel syndrome, which may be caused by diabetic


neuropathy, is usually treated with corticosteroids. Conclusions
Recent studies show that the administration of proges- Based on current findings, progesterone should be rec-
terone after acute back pain or anticancer drug-induced ognized as essential not only in reproductive fields, but
neuropathy (vincristine) (39) could improve neuro- as a potential tool for the management of many clinical
pathic pain by promoting myelin regeneration. More- conditions, including Alzheimer’s disease, cerebral
over, although estrogen therapy is associated with a edema, osteoporosis and diabetic neuropathy. A better
beneficial effect mediated by high-density lipoprotein understanding of the biological genomic and non-ge-
(HDL) increase, the addition of micronized proges- nomic receptor mechanisms could make it possible in
terone to estrogens in HRT has a negative impact on the near future to obtain a more comprehensive knowl-
HDL levels. edge of the safety and efficacy of this agent during hor-
mone replacement therapy (17 a-estradiol and natural
Progesterone and the mammary gland progesterone), with the aim of reducing breast cancer
risk.
During the luteal phase and, to a greater extent, in preg-
nancy, progesterone acts in synergy with estrogens, lead-
ing to a down-regulation of estrogen but not progestin References
receptors. The mitotic activity of breast epithelial cells is 1. Stefanick ML. Estrogens and progestins: background and
very high in the follicular phase but slows in the luteal history, trends in use, and guidelines and regimens
phase. This finding confirms the protective action of pro- approved by the US Food and Drug Administration. Am J
gesterone on breast tissue due to its inhibitory action on Med. 2005;118:64S–73S.
1 a-hydroxy steroid dehydrogenase and apoptosis (40). 2. Birch AJ. Hydro aromatic steroid hormones. Part I.
Several studies have been performed to evaluate HRT 10-Nortestosterone. J Chem. Soc., 1950, 367–368
safety in postmenopausal women, especially regarding the DOI: 10.1039/JR9500000367.
risk of developing breast cancer. The Women’s Health 3. Sandor T, Mehdi AZ. Steroids and evolution. In:
Initiative (WHI) study in 2002 showed an increased, Barrington EJW (ed). Hormones and Evolution. New
although not significant, risk of developing breast cancer York: Academic Press, 1979. pp. 1–72.
after HRT with conjugated estrogens and micronized pro- 4. Genazzani AR (ed). Endocrinologia ginecologica. Editeam,
gesterone. In 2008, data from the French E3N cohort Cento (FE), Italy. 2004;1–464.
5. Andersen JM, Dietschy JM. Relative importance of high
study were used to assess and compare the association
and low density lipoproteins in the regulation of
between different HRTs and breast cancer risk. During
cholesterol synthesis in the adrenal gland, ovary and testis
follow up, 2354 cases of invasive breast cancer occurred
in the rat. J Biol Chem. 1978;253:9024–6.
among 80 377 postmenopausal women. In that analysis
6. Brown MS, Goldstein JL. Receptor-mediated control of
the association of estrogen and different progestins in
cholesterol metabolism. Science. 1976;191:150–5.
HRT did not significantly increase breast cancer risk; 7. Amadori A, Cavallari C, Giacomucci E, Macrelli S,
however, the association of estrogen-micronized proges- Mastronuzzi G, Ucci N. (eds). Fisiologia della
terone and estrogen-dydrogesterone showed a trend Riproduzione. Bologna: CLUEB, 1994;1–192.
toward a lower risk (41). 8. Ryan KJ, Petro Z, Kaiser J. Steroid formation by isolated
In 2013, the results of the Kronos Early Estrogen and recombined granulosa and thecal cells. J Clin
Prevention Study (KEEPS) were published; in addition to Endocrinol Metab. 1968;28:355–60.
showing the beneficial effect of HRT on the cardiovascu- 9. Kobayashi M, Nakano R, Ooshima A.
lar system and neurocognitive sphere, this study did not Immunohistochemical localization of pituitary
detect a significant increase of breast cancer risk after gonadotropins and gonadal steroids confirms the “two-cell,
HRT with conjugated equine estrogen 0.45 mg/day or two-gonadotrophin” hypothesis of steroidogenesis in the
transdermal estradiol 50 lg/day, and micronized proges- human ovary. J Endocrinol. 1990;126:483–8.
terone (200 mg) administered for 12 days per month 10. Peluso JJ. Progesterone receptor membrane component 1
(42). and its role in ovarian follicle growth. Front Neurosci.
The REPLENISH trial is a phase 3 randomized placebo 2013;10:3389–95.
controlled study designed to evaluate the safety of a new 11. Frederick JL, Shimanuki T, diZerega GS. Initiation of
formulation of solubilized 17 a-estradiol and natural angiogenesis by human follicular fluid. Science.
micronized progesterone combined in a single gelatin 1984;224:389–90.
capsule opening a safety window between uterine activity 12. Revindranath N, Little-Ihrig L, Benyo DF, Zeleznik AJ.
Role of LH in the expression of cholesterol side-chain
and proliferative mammary gland (43).

14 ª 2015 Nordic Federation of Societies of Obstetrics and Gynecology, Acta Obstetricia et Gynecologica Scandinavica 94 (2015) 8–16
16000412, 2015, S161, Downloaded from https://obgyn.onlinelibrary.wiley.com/doi/10.1111/aogs.12771 by Nat Prov Indonesia, Wiley Online Library on [25/07/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Stefania Taraborrelli Physiology of progesterone

cleavage Cit P-450 and 3 a-hydroxy dehydrogenase antiapoptotic action in spontaneously immortalized
isomerase messenger ribonucleic acids in the primate granulosa cells. Endocrinol. 2004;145:3014–22.
corpus luteum. Endocrinol. 1992;131:2065–8. 27. Koensgen D, Mustea A, Klaman I, Sun P, Zafrakas M,
13. Filicori M, Butler JP, Crowley WF Jr. Neuroendocrine Lichtenegger W, et al. Expression analysis and RNA
regulation of the corpus luteum in the human. Evidences localization of PAI RBP1 (SERBP1) in epithelial ovarian
for pulsatile gonadotropin secretion. J Clin Endocrinol. cancer association with tumor progression. Gynecol Oncol.
1984;73:1638–47. 2007;107:266–73.
14. Lessey BA, Killam AP, Metzger DA, Haney AF, Greene GL, 28. Bolvin G, Anthoine-Terrier C, Morel G. Ultrasound of
McCarty KS Jr. Immunohistochemical analysis of human endogenous hormones and receptors in bone tissue: an
uterine estrogen and progesterone receptors throughout immunocytological approach in frozen sections. Micron.
the menstrual cycle. J Clin Metabol. 1988;67:334–8. 1994;25:15–27.
15. Satyaswaroop PG, Wartell DJ, Mortel R. Distribution of 29. Greenberg C, Kukreja SC, Bowser EN, Hargis GK, Henderson
progesterone receptor, estradiol dehydrogenase and 20 WJ, Williams GA. Effects of estradiol and progesterone on
alpha-dihydroprogesterone dehydrogenase activities in calcitonin secretion. Endocrinology. 1996;118:2594–8.
human endometrial glands and stroma: progestin 30. Delmas PD. A combination of low dose of 17 beta
induction of steroid dehydrogenase activities in vitro is estradiol and norethisterone acetato prevents bone loss and
restricted to the glandular epithelium. Endocrinology. normalizes bone turnover in postmenopausal women.
1982;111:743–9. Osteoporos Int. 2000;11:177–87.
16. The Writing Group for the PEPI Trial. Effects of hormone 31. Wren BG. Transdermal progesterone and its effects on
replacement therapy on endometrial histology in vasomotor symptoms, blood lipid levels, bone metabolic
postmenopausal women. The Postmenopausal Estrogen/ markers, moods, and quality of life for postmenopausal
Progestin Interventions (PEPI) Trial. JAMA. 1996;275:370–5. women. Menopause. 2003;10:13–8.
17. Csapo AL, Pukkinen MO. Effects of luteectomy and 32. Yao W, Dai W, Shahnazari M, Pham A, Chen Z, Chen H,
progesterone replacement in early pregnant patients. Am J et al. Inhibition of the progesterone nuclear receptor
Obstet Gynecol. 1973;115:759–64. during the bone linear growth phase increases peak bone
18. Schneider MA, Davies MC, Honour JW. The timing of mass in female mice. PLoS ONE. 2010;5:e11410.
placental competence in pregnancy after oocyte donation. 33. Goodman RL, Karsch FJ. Pulsatile secretion of LH:
Fertil Steril. 1993;59:1059–63. differential suppression by ovarian steroids. Endocrinology.
19. Parker MG, Christian M, White R. The nuclear receptor 1980;107:1286–90.
co-repressor RIP140 controls the expression of metabolic 34. Sayeed I. Progesterone is neuroprotective factor in
gene network. Biochem Soc Trans. 2006;34:1103–6. traumatic and ischemic brain injury. Prog Brain Res.
20. Lonard DM, Lanz RB, O’Malley BW. Nuclear receptor 2009;175:219–37.
coregulators and human disease. Endocr Rev. 2008;28:575– 35. Wang JM. Regeneration in a degenerating brain: potential
87. of allopregnagnolone as a neuroregenerative agent. Curr
21. Gellersen B, Fernandes MS, Brosens JJ. Non genomic Alzheimer Res. 2007;4:510–7.
progesterone actions in female reproduction. Human 36. Pinna G, Agis-Balboa RC, Pibiri F, Nelson M, Guidotti A,
Reprod Update. 2009;15:119–38. Costa E. Neurosteroid byosinthesis regulates sexually
22. Kastner P, Krust A, Turcotte B, Stropp U, Tora L, dimorphic fear and aggressive behavior in mice.
Gronemeyer H, et al. Two distinct estrogen-regulated Neurochem Res. 2008;33:1990–2007.
promoters generate transcripts endcoding the two 37. Ziomkiewicz A1, Pawlowski B, Ellison PT, Lipson SF,
functionally different human progesterone receptor forms Thune I, Jasienska G. Higher luteal progesterone is
A and B. EMBO J. 1990;9:1603–14. associated with low levels of premenstrual aggressive
23. Condon JC, Hardy DB, Kovaric K, Mendelson CR. Up- behavior and fatigue. Biol Psychol. 2012;91:376–82.
regulation of the progesterone receptor C isoform in 38. Kaore SN, Langade DK, Yadav VK, Sharma P, Thawani
laboring myometrium by activation of nuclear factor B VR, Sharma R. Novel actions of progesterone: what we
may contribute to the onset of labor through inhibition of know today and what will be the scenario in the future? J
PR function. Mol Endocrinol. 2006;20:764–75. Pharm Pharmacol. 2012;64:1040–62.
24. Fu X, Simoncini T. Non genomic sex steroid actions in the 39. Meyer L, Patte-Mensah C, Taleb O, Mensah-Nyagan AG.
vascular system. Semin Reprod Med. 2007;25:178–86. Cellular and functional evidence for a protective action of
25. Revelli A. Non genomic actions of steroid hormones in neurosteroids against vincristine chemotherapy-induced
reproductive tissues. Endocrin Rev. 1998;19:3–17. painful neuropathy. Cell Mol Life Sci. 2010;67:3017–34.
26. Peluso JJ, Pappalardo A, Fernandez G, Wu CA. 40. De Lignieres B. Estradiol and progesterone regulate the
Involvement of an unnamed protein, RDA288, in the proliferation of human breast epithelial cells. Fertil Steril.
mechanism through which progesterone mediates its 1998;69:963–9.

ª 2015 Nordic Federation of Societies of Obstetrics and Gynecology, Acta Obstetricia et Gynecologica Scandinavica 94 (2015) 8–16 15
16000412, 2015, S161, Downloaded from https://obgyn.onlinelibrary.wiley.com/doi/10.1111/aogs.12771 by Nat Prov Indonesia, Wiley Online Library on [25/07/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Physiology of progesterone Stefania Taraborrelli

41. Fournier A, Berrino F, Clavel-Chapelon F. Unequal risks 55. Peluso JJ, Fernandez G, Pappalardo A, White BA.
for breast cancer associated with different hormone Characterization of a putative membrane receptor for
replacement therapies: results from E3N cohort study. progesterone in rat granulosa cells. Biol Reprod.
Breast Cancer Res. 2008;107:103–11. 2001;65:94–101.
42. Wharton W, Gleason CE, Miller VM, Asthana S. Rationale 56. Bielefeldt K, Waite L, Abboud FM, Conklin JL.
and design of the KEEPS and the KEEPS Cognitive and Nongenomic effects of progesterone on human intestinal
affective sub study. Brain Res. 2013;1514:12–7. smooth muscle cells. Am J Physiol. 1996;271:G370–6.
43. Mirkin S, Amadio JM, Bernick BA, Pickar JH, Archer DF. 57. Barbagallo M, Dominguez LJ, Licata G, Shan J, Bing L,
17 b-estradiol and natural progesterone for menopausal Karpinski E, et al. Vascular effects of progesterone: role of
hormone therapy: REPLENISH phase 3 study design of cellular calcium regulation. Hypertension. 2001;37:142–7.
combination capsule and evidence review. Maturitas. 58. Rossato M, Nogara A, Merico M, Ferlin A, Foresta C.
2015;81:28–35. Identification of functional binding sites for progesterone in
44. Luconi M, Francavilla F, Porazzi I, Macerola B, Forti G, rat Leydig cell plasma membrane. Steroids. 1999;64:168–75.
Baldi E. Human spermatozoa as a model for studying 59. El-Hefnawy T, Huhtaniemi I. Progesterone can participate
membrane receptors mediating rapid nongenomic effects in downregulation of the luteinizing hormone receptor
of progesterone and estrogens. Steroids. 2004;69:553–9. gene expression and function in cultured murine Leydig
45. Blackmore PF, Neulen J, Lattanzio F, Beebe SJ. Cell cells. Mol Cell Endocrinol. 1998;137:127–38.
surface-binding sites for progesterone mediate calcium 60. Frye CA, Sumida K, Lydon JP, O’Malley BW, Pfaff DW.
uptake in human sperm. J Biol Chem. 1991;266:18655–9. Mid-aged and aged wild-type and progestin receptor
46. Kirkman-Brown JC, Bray C, Stewart PM, Barratt CL, knockout (PRKO) mice demonstrate rapid progesterone
Publicover SJ. Biphasic elevation of [Ca2+]i in individual and 3a,5a-THP-facilitated lordosis. Psychopharmacology.
human spermatozoa exposed to progesterone. Dev Biol. 2006;185:423–32.
2000;222:326–35. 61. Verikouki CH, Hatzoglou CH, Gourgoulianis KI,
47. Maller JL. The elusive progesterone receptor in Xenopus Molyvdas PA, Kallitsaris A, Messinis IE. Rapid effect of
oocytes. Proc Natl Acad Sci USA. 2001;98:8–10. progesterone on transepithelial resistance of human fetal
48. Bagowski CP, Myers JW, Ferrell JE Jr. The classical membranes: evidence for non-genomic action. Clin Exp
progesterone receptor associates with p42 MAPK and is Pharmacol Physiol. 2008;35:174–9.
involved in phosphatidylinositol 3-kinase signaling in 62. Fu XD, Flamini M, Sanchez AM, Goglia L, Giretti MS,
Xenopus oocytes. J Biol Chem. 2001;276:37708–14. Genazzani AR, et al. Progestogens regulate endothelial
49. Dosiou C, Hamilton AE, Pang Y, Overgaard MT, Tulac S, actin cytoskeleton and cell movement via the actin-binding
Dong J, et al. Expression of membrane progesterone protein moesin. Mol Hum Reprod. 2008a;14:225–34.
receptors on human T lymphocytes and Jurkat cells and 63. Fu XD, Giretti MS, Baldacci C, Garibaldi S, Flamini M,
activation of G-proteins by progesterone. J Endocrinol. Sanchez AM, et al. Extra-nuclear signaling of progesterone
2008;196:67–77. receptor to breast cancer cell movement and invasion
50. Ehring GR, Kerschbaum HH, Eder C, Neben AL, Fanger through the actin cytoskeleton. PLoS ONE. 2008b;3:e2790.
CM, Khoury RM, et al. A nongenomic mechanism for 64. Kaur P, Jodhka PK, Underwood WA, Bowles CA, de
progesterone-mediated immunosuppression: inhibition of Fiebre NC, de Fiebre CM, et al. Progesterone increases
K+ channels, Ca2+ signaling, and gene expression in T brain-derived neuroptrophic factor expression and protects
lymphocytes. J Exp Med. 1998;188:1593–602. against glutamate toxicity in a mitogen-activated protein
51. Bar J, Lahav J, Hod M, Ben-Rafael Z, Weinberger I, kinase- and phosphoinositide-3 kinase-dependent manner
Brosens J. Regulation of platelet aggregation and adenosine in cerebral cortical explants. J Neurosci Res.
triphosphate release in vitro by 17b-estradiol and 2007;85:2441–9.
medroxyprogesterone acetate in postmenopausal women. 65. Nilsen J, Brinton RD. Divergent impact of progesterone
Thromb Haemost. 2000;84:695–700. and medroxyprogesterone acetate (Provera) on nuclear
52. Blackmore PF, Fisher JF, Spilman CH, Bleasdale JE. Unusual mitogenactivated protein kinase signaling. Proc Natl Acad
steroid specificity of the cell surface progesterone receptor Sci USA. 2003;100:10506–11.
on human sperm. Mol Pharmacol. 1996;49:727–39. 66. Cai W, Zhu Y, Furuya K, Li Z, Sokabe M, Chen L. Two
53. Blackmore PF. Progesterone metabolites rapidly stimulate different molecular mechanisms underlying progesterone
calcium influx in human platelets by a src-dependent neuroprotection against ischemic brain damage.
pathway. Steroids. 2008;73:738–50. Neuropharmacology. 2008;55:127–38.
54. Peluso JJ, Pappalardo A. Progesterone regulates granulosa 67. Koulen P, Madry C, Duncan RS, Hwang JY, Nixon E,
cell viability through a protein kinase G-dependent McClung N, et al. Progesterone potentiates IP3-mediated
mechanism that may involve 14-3-3s. Biol Reprod. calcium signaling through Akt/PKB. Cell Physiol Biochem.
2004;71:1870–8. 2008;21:161–72.

16 ª 2015 Nordic Federation of Societies of Obstetrics and Gynecology, Acta Obstetricia et Gynecologica Scandinavica 94 (2015) 8–16

You might also like