Abstract

This mini-review summarizes key points from the Clark Sawin Memorial Lecture on the History of Estrogen delivered at Endo 2018 and focuses on the rationales and motivation leading to various discoveries and their clinical applications. During the classical period of antiquity, incisive clinical observations uncovered important findings; however, extensive anatomical dissections to solidify proof were generally lacking. Initiation of the experimental approach followed later, influenced by Claude Bernard’s treatise “An Introduction to the Study of Experimental Medicine.” With this approach, investigators began to explore the function of the ovaries and their “internal secretions” and, after intensive investigations for several years, purified various estrogens. Clinical therapies for hot flashes, osteoporosis, and dysmenorrhea were quickly developed and, later, methods of hormonal contraception. Sophisticated biochemical methods revealed the mechanisms of estrogen synthesis through the enzyme aromatase and, after discovery of the estrogen receptors, their specific biologic actions. Molecular techniques facilitated understanding of the specific transcriptional and translational events requiring estrogen. This body of knowledge led to methods to prevent and treat hormone-dependent neoplasms as well as a variety of other estrogen-related conditions. More recently, the role of estrogen in men was uncovered by prismatic examples of estrogen deficiency in male patients and by knockout of the estrogen receptor and aromatase in animals. As studies became more extensive, the effects of estrogen on nearly every organ were described. We conclude that the history of estrogen illustrates the role of intellectual reasoning, motivation, and serendipity in advancing knowledge about this important sex steroid.

This mini-review covers the salient points of the Clark Sawin Memorial Lecture delivered by the authors at the 2018 Endocrine Society annual meeting. Our knowledge about estrogen has been driven by astute observations of multiple investigators. The research findings have been rapidly translated into development of clinically applicable estrogen preparations. The discoveries have led to the birth control pill, prevention and treatment of breast cancer, menopausal hormone therapy, induction of puberty in hypogonadal girls, prevention and treatment of osteoporosis, and vaginal estrogen therapy, among others. Recent clinical and experimental data demonstrate important effects of estrogen on bone and reproductive function in men. With the involvement of a myriad of investigators in estrogen research, multiple controversies have arisen, some of which are still unresolved.

The Sawin presentation concentrated on the rationale for the various experiments, the data obtained, the controversies, and identification of the three types of investigators contributing to estrogen research (as pointed out by Fuller Albright). Albright, a giant in the field of endocrinology, quoted the US Supreme Court justice Oliver Wendell Holmes to define three categories of investigators as paraphrased succinctly: “One story intellects—fact collectors who have no aims beyond their facts; two story intellects who compare, reason and generalize using the labors of fact collectors as well as their own; three story intellects who idealize, imagine, predict—their best illumination comes from above through the skylight” (a phrase that the authors define as inspiration) (1).

Role of Historical Observations

In antiquity, the role of estrogens could only be inferred by astute observations both of external appearances and, later on, by anatomic dissection. The depiction of “King” Tutankhamun (1341 to 1323 bc) from a statue found in his tomb strongly suggests gynecomastia (Fig. 1) and represents an historical observation of estrogen excess in a male (2). Although some have suggested that this is merely artistic license, the similar representation of gynecomastia in his brother Smenkhkare, as well as his relatives Amenophis III and Akhenaten, leads us and others to think that this was indeed gynecomastia, suggesting to many the possibility of familial aromatase excess. Availability of Tutankhamun’s DNA should allow this to be diagnosed. Alternatively as suggested by Bernadine Paulshock, androgen insensitivity may have been responsible.

Photo of Tutankhamun taken by one of the authors (R.J.S.) at the Cairo Museum. Tutankhamun ruled from 1332 to 1323 bc. The statue depicts the appearance of gynecomastia, although some observers have speculated that this was only artistic license (2).
Figure 1.

Photo of Tutankhamun taken by one of the authors (R.J.S.) at the Cairo Museum. Tutankhamun ruled from 1332 to 1323 bc. The statue depicts the appearance of gynecomastia, although some observers have speculated that this was only artistic license (2).

Knowledge at the Height of the Roman Civilization

Physicians practicing in Rome often referred to the thoughts of Greek writers from their experience several centuries before. They knew that Aristotle (384–322 bc), for example, thought that only male semen was incorporated into the fetus and that the female played no role in the generative material (3). Soranus of Ephesus (ad 98–138), the most noted gynecologist at the time, contradicted Aristotle, in writing that both the male and female produce “seeds” necessary for conception (4–6). Galen (ad 129–200), the most accomplished of all medical researchers in antiquity and physician to the philosopher emperor Marcus Aurelius (author of The Meditations of Marcus Aurelius), appeared to agree with Soranus but not Aristotle (3, 7, 8). He observed female “testes” and concluded from these observations that the structures he saw corresponded to male testes and served the same purpose, namely production of semen (3).

Interestingly, Galen thought that menstruation represented a form of auto-phlebotomy and represented a means to eliminate unfavorable circulating humors, a concept not contradicted until several centuries later (3). Soranus commented on several aspects of female development. He noted a clear temporal association between menses and breast development and noted that these two events occur at approximately age 14 (6). This is of interest because it is generally agreed that the age of menarche has decreased from age 16 to 17 to age 13 during the past two centuries, a finding postulated to be due to increased fat deposition and leptin levels (9–12). This raises the question whether nutrition during the Roman civilization was better than two centuries ago in Western countries. The change in age of menstruation from ad 1800 to 1980 is thought to have resulted from the improvement in nutrition during that time period (9). As further evidence of the power of observation, Soranus also noted that masculine-appearing females and those exercising excessively failed to menstruate. He also commented on contraception, noting that blockade of the cervical os as an effective means of preventing conception was possible (6).

Post-Renaissance Period

Thomas Wharton (ad 1614–1673) was interested in ductless vs ductal glands. He was famous for naming Wharton’s duct, which connects the submandibular gland with the mouth. He thought, based on appearance, that the ovaries and testes are similar and that damage to either results in infertility (13). He reasoned on this basis, as did Soranus and Galen, that the ovary must produce sperm. Later, Augustin Nicolas Gendrin (ad 1796–1890) postulated that menstruation was related to ovulation, thus contradicting Galen’s theory of auto-phlebotomy (3, 7, 8, 14, 15).

Dawn of Experimental Era

In 1855, Claude Bernard, the father of experimental medicine, spearheaded the transition from clinical observation to experimentation (16). He developed the concept of internal secretion when observing that glucose released by the liver is transported by blood into many distant tissues. This observation led to a plethora of studies of internal secretion, basically involving three experimental steps: (i) removal of the gland with documentation of ensuing signs and symptoms, (ii) transplantation of the gland back into the body with demonstration of the reversal of signs and symptoms, and (iii) administration of the extract of the gland (organ therapy) in patients lacking that gland with the finding of reversal of signs and symptoms. With respect to the ovary, these steps were taken during a period of years.

Step 1

In the 1880s, Robert Battey developed the ability to perform oophorectomy safely in women (14, 17). This operation, called the Battey operation, became popular and was performed for multiple reasons, including dysmenorrhea and bleeding from myomata. After removal of the ovaries, patients developed hot flashes and vaginal atrophy, leading to the hypothesis that the ovary makes some type of substance that in its absence causes various symptoms (14, 17).

Step 2

In 1896, Emil Knauer from Vienna removed the ovaries from rabbits and observed uterine atrophy, which he could prevent by transplanting the ovary at a distant site, confirming the postulate of internal secretion by the ovaries (18–20).

Step 3

In 1897, Hubert Fosbery successfully used ovarian extracts to treat a patient with severe hot flashes (21). In his published report, he stated, “when at last the periods ceased, the patient was much troubled with frequent and violent flushings which at night in winter would wake her up, the face being in burning heat”; “I ordered 5-grain palatinoids of ovarian gland [a form of medicine capsule, as ground-up ovary tastes terrible] three times a day”; and “The flushings rapidly became less frequent and intense and were nearly cured by the time three dozen palatinoids were taken.” This description seemed rather convincing, although later it might have been attributed to a placebo effect (21).

History of Organ Therapy

The use of extracts of glands to treat patients became common in the later 19th and early 20th centuries. In the 1880s, Brown-Séquard (ad 1847–1894), thought to be the father of organ therapy, administered extracts of monkey testicles to patients with the concept that this would relieve symptoms due to specific hormone deficiencies (22). He administered these extracts to himself and reported increased strength and muscle mass, less fatigue, and an increase in mental faculties (23). He also suggested organ therapy for women whose ovaries had been removed. In response to Brown-Séquard’s wide influence, organ therapy then began to be used for many maladies, including mental disorders (24).

A “deep dive” into history revealed that Brown-Séquard was not the first to use organ therapy widely. In actuality, San Si Miao (ad 623–682) from China first administered the organ extracts of deer and sheep thyroid to patients with goiter and observed beneficial effects (Supplemental Fig. 1) (25, 26). He went from city to city expounding this therapy and demonstrating efficacy. From this and his publication of more than 40 volumes of medical information, he was then known as the “King of Medicine.” Later, in ad 1025, the Chinese prepared extracts of male and female urine, purportedly using powdered components to treat hypogonadism in men and dysmenorrhea in women, and other clinical disorders. The detailed methods describing preparation of these urine extracts is published in detail in the book The Genius of China and is summarized in Table 1 (27). What was actually contained in these extracts will remain unknown until the methods are replicated and analyzed with modern methods.

Table 1.

Methods of Preparing Urine Extracts to Treat Hypogonadism in Men, Dysmenorrhea in Women, and Other Clinical Disorders

StepsDescription
1Obtain 150 gallons of urine
2Heat to facilitate evaporation
3Seal and sublimate and obtain residue
4Grind to powder and make pill
5Administer five to seven pills with wine or warm soup
6Male urine used for hypogonadism, impotence, and beard growth
7Female urine used for dysmenorrhea
StepsDescription
1Obtain 150 gallons of urine
2Heat to facilitate evaporation
3Seal and sublimate and obtain residue
4Grind to powder and make pill
5Administer five to seven pills with wine or warm soup
6Male urine used for hypogonadism, impotence, and beard growth
7Female urine used for dysmenorrhea

Data from Temple R. The Genius of China: 3,000 Years of Science, Discovery, and Invention. Prion, 1983.

Table 1.

Methods of Preparing Urine Extracts to Treat Hypogonadism in Men, Dysmenorrhea in Women, and Other Clinical Disorders

StepsDescription
1Obtain 150 gallons of urine
2Heat to facilitate evaporation
3Seal and sublimate and obtain residue
4Grind to powder and make pill
5Administer five to seven pills with wine or warm soup
6Male urine used for hypogonadism, impotence, and beard growth
7Female urine used for dysmenorrhea
StepsDescription
1Obtain 150 gallons of urine
2Heat to facilitate evaporation
3Seal and sublimate and obtain residue
4Grind to powder and make pill
5Administer five to seven pills with wine or warm soup
6Male urine used for hypogonadism, impotence, and beard growth
7Female urine used for dysmenorrhea

Data from Temple R. The Genius of China: 3,000 Years of Science, Discovery, and Invention. Prion, 1983.

Therapeutic Innovation Based on Evolving Concepts

As the concepts regarding the function of the ovaries evolved, therapies were designed based on the current knowledge. In 1896, Sir George Beatson, a surgeon, described the first effective hormonal ablative therapy for treatment of breast cancer (28–30). He based his rationale on an experience “moonlighting” on a farm where he was the physician for the owner. There he learned several facts about lactation and began to study this phenomenon. Based on his knowledge of histology, he appreciated similarities between the tissue appearance of lactational changes, characterized by increasing breast proliferation and the same phenomenon in histologic sections of breast cancer. Also, he knew that oophorectomy prolonged the time of lactation in cows, a practice common in Australia at that time. From these observations, he postulated a regulatory role of the ovary on benign and malignant breast and sought to apply this concept clinically. With seemingly great courage, he decided to remove the ovaries surgically in premenopausal women with breast cancer. Notably, he demonstrated both partial and complete, temporary remissions. This work initiated the field of surgical ablation of endocrine glands as treatment of hormone-dependent breast cancer. To the authors of this manuscript, this represented “ three story thinking,” primarily with inspiration as to the rationale.

Purification of Estrogen

A major accomplishment at the time was the purification of estrogen, a feat primarily involving three key investigators, Edgar Allen, Edward Doisy, and Adolph Butenandt. In 1923, Edgar Allen, a reproductive physiologist, was studying the role of follicular fluids obtained from the ovary of sows on uterine weight, vaginal maturation, and sexual receptivity (31, 32). A serendipitous circumstance leading to a key collaboration came from the fact that Allen had no car. Taking advantage of a friendship developed while playing on a faculty baseball team, he rode to work in St. Louis in the Model T Ford of Edward Doisy, an organic biochemist and fellow baseball enthusiast (33). This allowed extensive conversations leading to the appreciation of the potential for a synergistic, scientific relationship and commencement of work together—Allen provided follicular fluid from sows and Doisy purified the estrogenic activity. The biologic endpoint, uterine weight, was later called the Allen-Doisy test.

Progress was slow until Selmar Ascheim and Bernard Zondek, working in Germany, demonstrated large amounts of estrogenic activity in the urine of pregnant women (34). With this biological material as a source, Doisy crystallized estrone and presented this finding at the 13th International Physiologic Congress in Boston in 1929 (35, 36). Fuller Albright observing the presentation commented, “ The potency of these crystals is so great that one gram could restore the sex cycle in more than nine million rats” (1). Likely, Albright began to think about the implications, an approach characterizing him as a “three story investigator.” After discovering the crystals, Doisy went home and told his wife that they were going to be rich. On second thought, he gave the two resulting patents (both awarded on 24 July 1934) to St. Louis University, which has used the monies in an established foundation to this day to support the biochemistry department in the medical school.

Groundbreaking research findings often occur simultaneously in two completely independent laboratories. The German investigator Adolph Butenandt had simultaneously purified and crystalized estrone when he heard about Doisy’s talk from a colleague who had attended the Boston meeting. This led Butenandt to rush into print several months before Doisy’s manuscript was published (37). Ironically, Butenandt (along with Leopold Ruzincka) won the Noble Prize for this work (as well as for the crystallization of testosterone), whereas Doisy had to wait a decade to win his Nobel Prize for the purification of vitamin K. Somewhat later, estriol and estradiol were isolated and purified (38, 39). As expected, the problem of fully characterizing the structure of estrogens required major feats of organic chemical investigation, leading to controversies among investigators. A major one was whether estrogens contained three or four cyclohexane rings, a controversy illustrated by a photograph of Butenandt displaying four fingers, which aptly conveyed his opinion [see Fig. 3 in Simpson and Santen (40)].

Early Clinical Applications

Within 5 years, several estrogen preparations were commercially available from multiple pharmaceutical companies that recognized the clinical and commercial benefits of estrogen. The various products included theelin, progynon, emmenin, oestroform, folliculin, and amniotin. The ready availability of these agents facilitated rapid clinical advances. In 2 years, Albright demonstrated that estrogen blocked hot flashes; in 5 years, bone loss; and in 6 years, ovulation (41–45). Of interest was that only one patient needed to be studied with cyclic on–off therapy with estrin (preparation of estrogen based on the Doisy formula) to convincingly demonstrate inhibition of hot flashes (43) (Fig. 2). Estrin also provided relief of dysmenorrhea (45). The observation that estrogen markedly increased the amount of bone in male pigeons (46) (Fig. 3) stimulated Albright to show that estrogens prevent bone loss in postmenopausal women (44, 47, 48) (Fig. 4). This emergence of these new findings led Albright to establish a clinic specifically related to reproduction. In this clinic, Albright demonstrated blockade of ovulation with administration of estrogen during the early follicular phase of the menstrual cycle (42, 43).

Treatment of a patient with estrin as shown by the hatched bars. The number of hot flashes per week are shown on the vertical axis. [Reproduced with permission from Albright F. Studies on ovarian dysfunction III: the menopause. Endocrinology 1936;20:24–39.]
Figure 2.

Treatment of a patient with estrin as shown by the hatched bars. The number of hot flashes per week are shown on the vertical axis. [Reproduced with permission from Albright F. Studies on ovarian dysfunction III: the menopause. Endocrinology 1936;20:24–39.]

The femoral bone from an untreated male pigeon is shown on the left and after 36 days of estrogen on the right (46). [Reproduced with permission from Pfeiffer CA, Gardner WU. Skeletal changes and blood calcium level in pigeons receiving estrogens. Endocrinology 1938;23:485–491.]
Figure 3.

The femoral bone from an untreated male pigeon is shown on the left and after 36 days of estrogen on the right (46). [Reproduced with permission from Pfeiffer CA, Gardner WU. Skeletal changes and blood calcium level in pigeons receiving estrogens. Endocrinology 1938;23:485–491.]

The vertical axis shows the height of patients before starting estrogen therapy (left of the vertical line) and after starting hormone therapy (right of the vertical line). This clearly shows that estrogen prevents loss of height in postmenopausal women (48). [Reproduced with permission for electronic publication only from Wallach S, Henneman PH. Prolonged estrogen therapy in postmenopausal women. JAMA 1959;171:1637–1642.]
Figure 4.

The vertical axis shows the height of patients before starting estrogen therapy (left of the vertical line) and after starting hormone therapy (right of the vertical line). This clearly shows that estrogen prevents loss of height in postmenopausal women (48). [Reproduced with permission for electronic publication only from Wallach S, Henneman PH. Prolonged estrogen therapy in postmenopausal women. JAMA 1959;171:1637–1642.]

Physiology of Breast Cancer

From George Beatson’s work, the role of estrogen on breast cancer growth was known, but an animal model was needed to study the precise physiologic mechanisms. Charles Huggins had won the Nobel Prize for demonstrating that castration in men could cause prostate tumor regression (49). Surprisingly, late in his career he developed the first animal model of hormone-dependent breast cancer. Motivation is often an interesting component of research progress (50–54). One of the coauthors (R.J.S.) had the opportunity to ask Huggins why he embarked upon the new area of research late in his career. He responded “The Ben May laboratory had no operating room and I had to take the dogs to Northwestern for the orchiectomies in my wife’s station wagon. One day she said, “Charlie, your dogs are no longer going to befoul my station wagon and you had better find something else to work on.” He chose to develop the dimethylbenzathracene (DMBA) breast cancer model, which allowed much information to be obtained about the hormone responsiveness of this common neoplasm in women.

Development of the Birth Control Pill

Several key steps led up to this monumental achievement (55, 56). In 1949, Russell Marker found a method to markedly reduce the cost of synthesizing progesterone by identifying precursors in a specific Mexican yam. He spent several months testing more than 100 yams, leading to the identification of the one with the highest level of diosgenin tested (55, 57, 58). He used this as a starting material for the synthesis of progesterone, which resulted in a lowering of the price from US$200 per gram to US$5 (55). Gregory Pincus and Min-Chueh Chang then demonstrated blockade of ovulation in animals with progestins. Clinical trials in Puerto Rico, involving several individuals, including John Rock, demonstrated the efficacy of contraception in women (55, 56). The background for these studies was that Margaret Sanger, founder of the “National Birth Control League,” later named Planned Parenthood, was well ahead of her time and strongly encouraged contraceptive research in the early 1920s. A surprisingly current quote from her at that time stated that “No woman can call herself free who does not control her own body.” The extensive research required a substantial amount of funding. Katharine Dexter McCormick, inheritor of the McCormick fortune from the McCormick Reaper, contributed nearly one-third of the money required but notably also imparted intensive intellectual input into birth control pill research (55).

John Rock, a staunch Roman Catholic obstetrician/gynecologist from Boston, thought that his church would approve the birth control pill, but this did not happen. A little known fact is the history of the Catholic Church’s deliberations on the morality of the birth control pill. A large and distinguished panel was convened by Pope John XXIII to consider whether “artificial birth control” was moral. The majority report of more than 60 experts considered it moral and their report was leaked to The New York Times [“An analysis of the majority report ‘Responsible Parenthood’ and its recommendations of abortion, sterilization, and contraception” by Richard J. Fehring (59)]. John XXIII died and his successor, Pope Paul VI, accepted the minority opinion of no more than 15 panel members and declared oral contraceptives to be morally unacceptable. Interestingly, most Catholic women in the United States approve of use of oral contraceptives despite church teaching.

Several outcomes ensued after approval of Enovid, the first oral contraceptive in 1960. Laws were changed to legalize contraceptives. This represented the first practical ability of families to determine the number of children desired and superseded the use of condoms and the “rhythm method,” derided by some as a form of Russian roulette. Availability of birth control pills led to substantial changes in the role of women in society and the workplace and putatively was one of the most powerful influences on several social aspects, including sexual behavior, gender roles in society, and family economics (60).

Discovery of the Estrogen Receptor

Novel techniques led to initial studies involving radioisotopes of high specific activity, namely tritium and carbon-14, which were used to label steroids such as estradiol.

Elwood Jensen, another “three story investigator,” first identified estrogen receptors (ERs) utilizing this methodology. He administered labeled estradiol to castrate rats and demonstrated specific uptake into the uterus and vagina (61) (Fig. 5). He then demonstrated that ERs in women could be used to predict which breast tumors were hormone-dependent and would respond to hormone therapy (62). As a mentor, he stimulated his mentees to purify the ER and study its detailed molecular mechanisms.

Administration of tritiated estradiol to oophorectomized rats. The disintegrations per minute (DPM) of isotope per milligram of dry tissue are shown on the vertical axis and time is shown on the horizontal axis (61). The red arrow on the left points out the peak concentration of isotope in the uterine tissue at 1.5 h, and the red arrow on the right the defection point in the decline at the 6-h time point. [Reproduced with permission for electronic publication only from Jensen EV. On the mechanism of estrogen action. Perspectives in Biology and Medicine 1962;6:47–59.]
Figure 5.

Administration of tritiated estradiol to oophorectomized rats. The disintegrations per minute (DPM) of isotope per milligram of dry tissue are shown on the vertical axis and time is shown on the horizontal axis (61). The red arrow on the left points out the peak concentration of isotope in the uterine tissue at 1.5 h, and the red arrow on the right the defection point in the decline at the 6-h time point. [Reproduced with permission for electronic publication only from Jensen EV. On the mechanism of estrogen action. Perspectives in Biology and Medicine 1962;6:47–59.]

Aromatase

Discovery

The aromatization of androstenedione to estrone and testosterone to estradiol was first suggested by Bernard Zondek in 1934 (34, 63). Later, Steinach and Kun “injected large quantities of male hormone (testosterone propionate) into men and quantitated the excretion of estrogenic substances in the morning urine” (64). The finding that rat units of excretion increased from 0 to 36 to a maximum of 1200 confirmed that androgens served as precursors for the product estrogens, a relationship suggesting the presence of aromatization. As often occurs in science, a controversy arose as to the naming of the enzyme. Some investigators called the enzyme “estrogen synthase” and others “aromatase,” but in the long run, aromatase won out (65).

Major biochemical studies were initiated in the 1960s. Ken Ryan studied and characterized placental aromatase and Harry Brodie and others the enzymatic steps involved (66–73). The initial two hydroxylation steps involved, steps 1 and 2, are generally agreed to involve sequential hydroxylations on the C19 methyl group. The third hydroxylation step remained elusive for more than two decades. Notably, recent studies from Guengerich’s group have shed additional light on this issue (74). As shown in Supplemental Fig. 2 (26, 74), he suggested two possibilities for step 3 and outlined the specific interactions necessary. This synthesis pathway highlights how one enzyme can remarkably catalyze three separate steps.

Purification

The tools necessary for protein purification in the 1980s involved multiple steps using sequential chromatographic columns and serial eluants with measurement of enzyme activity in the various fractions. The tritiated water assay, which used placental microsomal aromatase, developed earlier by Siiteri and Thompson (75–78), facilitated these studies by considerably simplifying the enzyme activity assay and reducing the need for thin layer chromatography. The overall process of purification was technically difficult, time-consuming, and costly. Nonetheless, Yoshio Osawa, Peter Hall, Larry Vickery, Frank Bellino, Norio Muto, Marrku Pasanen, Olavi Pelkonen, Evan Simpson, and Carole Mendelson purified the human aromatase cytochrome P450 protein from human placental microsomes (79–84). Final demonstration of activity required the confirmation of the tritiated water assay with thin layer chromatographic demonstration of the conversion of androstenedione to estrogen, crystallization, and then recrystallization. When aromatase was originally studied in the early 1950s, it was logical to think that this reaction required three separate enzymes. Conceptually, it was difficult to imagine that one enzyme could cause hydroxylation of three separate sites and the double bond structure in the A ring. Nonetheless, the purified enzyme could clearly catalyze these disparate steps, a finding considered by all investigators to be definitive.

During this period, the techniques of recombinant DNA revolutionized the ease of determination of exact protein structures, both by considerably simplifying the process and by the sensitivity and specificity of the methods. Availability of purified protein preparations of aromatase led to the generation of both polyclonal and monoclonal antibodies that could then be used for analyzing clones. The laboratories of Toda and Shizuta and Evans, Simpson, and Mendelson obtained clones complementary to aromatase transcripts. Expression libraries, and particularly one using the λgt11 phage, enabled isolation of a partial cDNA clone lacking the 5′ end of the cDNA but containing the heme binding region. Simpson and Mendelson carried out these studies and extended their findings using the 5′ rapid amplification of cDNA end technique (85) to identify the entire cDNA and 503 amino acid sequences. As biologic studies had identified aromatase in a number of species (75), a series of investigators used similar molecular biological methodology to identify aromatase sequences from numerous species and to compare and contrast the unique structural components of each (85–92).

Three-dimensional structure

As X-ray analysis of crystal structure provided a powerful tool to determine specific mechanisms for enzymatic activity, investigators in the aromatase field worked very hard for at least two decades to obtain crystals of this protein. They found this task to be problematic owing to the fact that the protein was membrane bound, relatively low in amount, and contained elements such as the heme configuration that were unstable during purification. As a means to overcome these obstacles, a number of groups sought to model the three-dimensional structure of the aromatase protein based on the known structures of other cytochrome P450 species (93–95). Promising candidates, known at that time, were soluble prokaryotic, cytochrome P450 species. Modeling attempts used these structures, which, however, had low sequence homology to human aromatase (96, 97). As the field evolved, the molecular biological tools enabled study of a wide variety of structures of mammalian microsomal P450 species that served as templates for model building (96–98). This approach, and particularly that used by Shiuan Chen and his group (98), proved to be remarkably predictive of later studies using human aromatase when it was finally crystalized (99).

In some instances in science, older, time-proven, but quite difficult techniques prove superior to newer, elegant molecular methodologic methods. This principle characterized the final crystallization of human aromatase. Starting with the original methodology of Yoshio Osawa (81), the group of Debashis Ghosh in Buffalo, New York, used protein chemistry methods to purify aromatase from human placenta in quantity and quality allowing crystallization. This process overall encompassed more than 20 years.

They then used this to obtain crystals that allowed characterization of the actual three-dimensional structure of human aromatase by X-ray spectroscopy (99) (Fig. 6). When all is taken into consideration, this would appear to be a remarkable achievement.

Close-up of the docking site for androstenedione in the structure of the aromatase enzyme obtained from analysis of aromatase crystals. [Reproduced with permission for electronic publication only from Ghosh D, Griswold J, Erman M, Pangborn W. X-ray structure of human aromatase reveals an androgen-specific active site. J Steroid Biochem Mol Biol 2010;118(4–5):197–202.]
Figure 6.

Close-up of the docking site for androstenedione in the structure of the aromatase enzyme obtained from analysis of aromatase crystals. [Reproduced with permission for electronic publication only from Ghosh D, Griswold J, Erman M, Pangborn W. X-ray structure of human aromatase reveals an androgen-specific active site. J Steroid Biochem Mol Biol 2010;118(4–5):197–202.]

Genomic Structure

Exons I through X

The next step was to determine the sequence of the human aromatase gene. This was achieved using the aromatase cDNA sequences as probes for human genomic libraries. In this way, the exonic sequence together with the flanking sequences of the human gene were obtained (84, 92–96). The sequence contained nine coding exons, which were named by the Dallas group II to X, with the heme-binding region present in exon X. This region was ∼30 kb in length. The first exon turned out to be upstream of the translational start site and moreover was different for each tissue site of expression. Namely, the placenta, ovary, and adipose tissue each exhibits a unique exon I (100–102). These are spliced into a common 3′ splice junction upstream of the start of translation in a tissue-specific fashion such that the sequence of the translated protein is always identical (Supplemental Fig. 3) (26). This was quite unexpected and important because the use of these untranslated first exons is directed by a number of tissue-specific promoters in a tissue-specific fashion. Because each of these promoters is uniquely regulated by a different cohort of trans-acting elements, this determines the tissue-specific regulation of aromatase expression and hence of estrogen biosynthesis.

When the sequence of the aromatase gene was matched to the published sequence of the human genome, there was a missing region between the most distant promoter, namely that of the placenta, and the remaining sequence. This alignment with genomic sequences enabled the gap to be closed by Bulun et al. (103) to reveal that the complete structure encompasses a span of ∼123 kb. Within the P450 superfamily, aromatase is designated as family 19, with the human gene as CYP19A, and the mouse gene as Cyp 19A.

Promoter regulation

At the time, this type of regulation was poorly appreciated. As indicated above, tissue specificity was enabled by the presence of tissue-specific promoters upstream of the various first exons, which then stimulated the transcription of the aromatase message contained in exons II through X. Thus, for example, upstream of the placenta-specific first exon is a promoter region (promoter I.1) that is regulated by multiple factors as shown in Supplemental Fig. 4 (26). Notably, however, this first exon is located some 90 kb upstream of the translational start site and the entire intervening sequence is spliced out during (100) processing of the placental aromatase transcript. The specific processes differ in various tissues. For example, in the ovarian tissue (101, 102), the first exon (exon II) is proximal to the translational start site and its promoter.

In the ovary, promoter II contains two cAMP response elements and hence is regulated by cAMP through actions of FSH. Aromatase expression in adipose tissue is recognized to be present largely in the preadipocytes or stromal cells rather than the lipid-laden adipocytes. In these cells, aromatase is regulated primarily by another distal promoter (promoter I.4) that is regulated by class 1 cytokines such as IL-6 and also by TNFα. However, transcripts from the ovarian promoter II are also present in adipose stromal cells, and these become dominant in cancer-associated fibroblasts surrounding a breast tumor due to the action of prostaglandin E2, probably produced largely by the tumorous epithelium (Supplemental Fig. 5) (75). The production of estrogen within cells and its binding to the estrogen in the same cells is an example of intracrinology (Supplemental Fig. 5) (26).

[The reader should note that two separate terminologies exist for alternative exons I as proposed by Mahendroo et al. (101) and Means et al. (102) (i.e., exons I.1, I.2, I.3, I.4, 2a) and by Harada et al. (100) (i.e., exons 1a, 1b, 1c, 1d, 1f, 2a).]

ER-Mediated Functions

ERα and ERβ

Both receptors are members of the nuclear receptor superfamily and contain six functional domains: amino terminal (A/B domain), DNA-binding C domain, hinge region (d domain), ligand-binding (E) domain, and carboxyl-terminal F domain (104–106). The A/B domain contains transcription activation function 1, and the ligand-binding domain contains transcription activation function 2 (104, 107). Binding of estradiol to both ERs changes the conformation of a critical portion of these receptors, helix 12, which exposes protein surfaces where both coactivators and corepressors can bind. These proteins can amplify or reduce the rate of transcription induced by binding to the estrogen ligand. Both ERα and ERβ contain a high degree of homology in the C and E domains but are divergent in the A/B, D, and F domains (104, 105). Alternative splicing and utilization of divergent start sites results in receptors of varying size. Additionally, breast cancers treated with hormone therapy develop ERα mutations that act in a ligand-independent fashion (108).

ERα levels are highest in the uterus and pituitary and in lesser concentrations in the liver, hypothalamus, bone, mammary gland, cervix, and vagina. ERβ levels are highest in the ovary (exclusively in the granulosa cells), lung, and prostate (109). Extensive knockout and knock-in experiments in mice have elucidated the specific physiologic roles of each receptor. As examples, ERβ is necessary for the proper differentiation of ovarian granulosa cells and efficient ovulation, whereas ERα is important for the function of the uterus (104, 110) and development of the mammary gland.

Sites of initiation of receptor-mediated events

The ER resides in the perimembranous region and can initiate events at that location. After synthesis, the ERs primarily localize to the nucleus due to a strong specific nuclear localization site originating in the D domain. ERα also is reported to be present in the mitochondria where it is involved in regulation of reactive oxygen species and apoptosis (111–113). Current concepts hold that events initiated at or near the cell membrane are integrated with mechanisms involving nuclear initiation (112, 114–127).

Nuclear-initiated events

Both ERα and ERβ can act in the nucleus via three separate mechanisms: (i) direct binding to estrogen response elements, which classically consist of 5GGTCAnnnTGACC palindromes or minor variants of this DNA sequence; (ii) tethering to AP-1 and SP-1, which in turn have their own response elements, or to activated MAPK; and (iii) ligand-independent receptor activation, which is thought to be possible when specific serines in the receptor structure are phosphorylated. Dimerization of two ER monomers is necessary for functionality. Chromatin immunoprecipitation sequencing studies have shown that the ER binds to 5000 DNA sites in the absence of ligand and 17,000 sites when ligand is present (128). Activation of transcription involves RNA polymerase II, which is only activated when ligand is present. This process involves the pioneering factor FOXA1, which causes chromatin remodeling and opens up sites for ER binding to the estrogen response elements (129). Coactivators and corepressors are important modulators of this process, as are complex enhancer elements. A simplified diagram of these events is illustrated in Supplemental Fig. 6 (26).

A myriad of investigators contributed to identifying each component in this complex fabric of functional activity. The details of each of these steps is beyond the scope of this minireview. However, to name just a few studies and investigators, Geoff Green, in conjunction with Pierre Chambon, and others cloned ERα and later crystalized this protein (Supplemental Fig. 7) (26, 130); Kuiper et al. (127) cloned ERβ; and multiple investigators studied genomic actions [see key manuscripts and reviews by Ken Korach, Donald McDonnell, John and Benita Katzenellenbogan, Serdar Bulun, Jack Gorski, Bert O’Malley, and many others (omitted because of space constraints)] (103, 104, 109, 115, 121, 125, 130–161).

Membrane-initiated actions

Pietras and Szego (162) first demonstrated membrane effects of estrogen and published these data in Nature in 1977. These studies were originally considered controversial and did not stimulate further investigations in this area. Two decades later, Ellis Levin and others began to explore this area intensively (112, 116–118). Studies during the past decade have clearly demonstrated a role for ERs residing near or in the plasma membrane and a role for ERα in the mitochondria (111, 113, 121, 136, 145, 162–174). Another estradiol-binding protein, GP 30, now called GPER1, also mediates multiple functions in various tissues (123, 173).

As a key issue related to the mechanism whereby ERα was directed to the plasma membrane, Razandi et al. (174) demonstrated that cytosolic ER is palmitoylated, which allows localization in or near the plasma membrane. Palmitoylation at cysteine 447 provides the mechanism for transport to the membrane where it binds to caveolin. Relevant studies then began examining the downstream signaling initiated by membrane ERα, which was found to be quite complex. Steps included rapid activation of several different components: IGF-1, epidermal growth factor, p21, Raf, MAPK, AKT, protein kinase C, release of nitric oxide, stimulation of prolactin secretion, and alteration of calcium and maxi-K channels (164). A marker of membrane signaling used by most investigators initially was MAPK, which is activated within 5 minutes of exposure to estradiol (164, 175, 176). Use of estradiol-linked dendrimer conjugates that could not enter the nucleus to activate MAPK (115, 153, 177) confirmed that this action was not nuclear. An important finding from these studies was that membrane-initiated events could ultimately result in regulation of transcription in the nucleus. Activation of MAPK, an event occurring at or near the membrane, resulted in ERα/MAPK complexes that entered the nucleus and bound to specific DNA response elements. There, activated MAPK can phosphorylate SRC3, RIP140, p300, and CREB1 (115, 177–179). The demonstration of ERα/MAPK complexes on DNA as shown by chromatin immunoprecipitation assays provided conclusive proof of this membrane to nuclear DNA pathway (115, 177). The signaling pathways that transduce the rapid effects of estradiol are complex and differ according to cell type.

Mitochondrial function

Mitochondria contain both ERα and ERβ. Estradiol can augment several mitochondrial DNA-encoded RNAs (113). Through ERβ, estrogen simulates manganese superoxide dismutase and reduces damage from reactive oxygen species. These actions lead to a reduction of apoptosis. An extensive literature has now been developed that identifies multiple effects of both ERα and ERβ on mitochondrial function (112, 113).

Estrogen acts on both sexes

Two prismatic cases clearly demonstrated the role of estrogen in men. Smith et al. (147) reported a male patient with a nonfunctional ERα, and Maffei et al. (180) reported a man with nonfunctioning aromatase. Both had osteopenia with genu valgum and continued to grow into their late 20s, lacking closure of their long bone epiphyses (Fig. 7). These findings indicated the role of estrogen in epiphyseal closure and bone maintenance in men. Treatment in the aromatase-deficient male demonstrated that estrogen regulates lipids, enhances insulin sensitivity, lowers glucose, and normalizes liver function, all important metabolic and hepatic effects (Supplemental Figs. 8 and 9) (26). Further information was gained using genetically altered mice with ERα/β and aromatase knockout or knock-in conducted by Ken Korach, Evan Simpson, Matti Poutanen, and Raj Tekmal and their respective groups (75, 125, 181–186). These studies demonstrated a role of estrogen in spermatogenesis, sexual intromission, ejaculation, and maintenance of bone density. Many of these effects were also demonstrated by Finkelstein et al. (187) who conducted add-back experiments in men given GnRH agonists to suppress androgen and estrogen levels.

The growth curve over time in a patient with aromatase deficiency (left panel) and an X-ray of the hand (right panel) in the same patient at age 24 to demonstrate the lack of closure of the epiphyses. The lack of closed epiphyses is characteristic of a 14 year old and thus the term, bone age of 14 is used (180). [Reproduced with permission from Smith EP, Boyd J, Frank GR, et al. Estrogen resistance caused by a mutation in the estrogen-receptor gene in a man. N Engl J Med 1994;331(16):1057.]
Figure 7.

The growth curve over time in a patient with aromatase deficiency (left panel) and an X-ray of the hand (right panel) in the same patient at age 24 to demonstrate the lack of closure of the epiphyses. The lack of closed epiphyses is characteristic of a 14 year old and thus the term, bone age of 14 is used (180). [Reproduced with permission from Smith EP, Boyd J, Frank GR, et al. Estrogen resistance caused by a mutation in the estrogen-receptor gene in a man. N Engl J Med 1994;331(16):1057.]

Clinical Studies

Early metabolic studies

A number of investigators began to extensively study the metabolism of various steroids using radioisotope kinetic studies in the late 1950s and 1960s. This provided the ideal methodology to examine the levels of aromatase activity under various clinical circumstances. Using these techniques, Pentti Siiteri and Paul MacDonald (188) studied the clinical expression of aromatase in patients as a function of both weight and age (Fig. 8). They demonstrated nearly a 10-fold increase in aromatase when comparing 90-pound subjects with 250- to 300-pound men and women. These results clearly showed that most of this estrogen was coming from the adipose tissue rather than the gonads and also demonstrated that extragonadal aromatization increased substantially as a function of increasing age (188).

The relationship of the percentage conversion of testosterone to estradiol under steady-state conditions (vertical axis) to body weight in pounds (lbs) in postmenopausal women. Correlation coefficient, 0.74 (188). [Reproduced with permission from Siiteri PK, MacDonald PC. Role of extraglandular estrogen in human endocrinology. In: Greep RO, Astwood EB, eds. Handbook of Physiology. Washington, DC: American Physiologic Society, 1973:615–629.]
Figure 8.

The relationship of the percentage conversion of testosterone to estradiol under steady-state conditions (vertical axis) to body weight in pounds (lbs) in postmenopausal women. Correlation coefficient, 0.74 (188). [Reproduced with permission from Siiteri PK, MacDonald PC. Role of extraglandular estrogen in human endocrinology. In: Greep RO, Astwood EB, eds. Handbook of Physiology. Washington, DC: American Physiologic Society, 1973:615–629.]

Clinical implications of estrogen physiology

Animal models of breast cancer and prior observations with surgical oophorectomy and adrenalectomy in breast cancer patients suggested that an antiestrogen might also be effective treatment (49, 189–191). This suggested the use of tamoxifen in patients with breast cancer. The first clinical trial was reported by Cole et al. (192) in 1971 at the Christie Hospital in Manchester, UK, demonstrating a 22% response rate in 46 patients receiving 10 mg daily of tamoxifen. A study in 1973 reported a 40% response in 35 women receiving 20 mg daily (193). These results compared favorably with non–head-to-head studies in women treated with ablative surgery (oophorectomy or adrenalectomy) that were routine at that time and strongly suggested the efficacy of medical, antiestrogen therapy (194). Later extensive studies by Craig Jordan and associates identified a unique property of tamoxifen, that is, that it exerts antiestrogenic effects on breast but estrogenic effects on uterus and bone. These findings led to the coining of the term SERM (selective ER modulator) (195).

Several investigators postulated that inhibition of adrenal steroid synthesis would be as effective as tamoxifen as treatment of breast cancer in postmenopausal women. Accordingly, the nonsteroidal inhibitor aminoglutethimide (then thought to be a cholesterol side chain-inhibitor) was studied by Robert Cash, Kenneth Gale, “Dickie” Newsome, and Richard Santen with demonstration of antitumor efficacy (75, 196–201). Later, stimulated by the advice of Pentti Siiteri, biochemical studies were undertaken that demonstrated that aminoglutethimide was an effective aromatase inhibitor (75–78, 188). The use of isotopic kinetic technology allowed demonstration in 1978 of a 95% reduction of aromatase with aminoglutethimide (Supplemental Fig. 10) (26), which represented its major mechanism of action (200).

Angela Brodie and her husband, Harry Brodie, focused on development of steroidal aromatase inhibitors. After preclinical studies were completed, Angela Brodie and her collaborators demonstrated the clinical efficacy of the steroidal aromatase inhibitor 4-OH-androstenedione (75, 202, 203) (Supplemental Fig. 11) (26). Full details of the history of development of aromatase inhibitors are detailed in another publication by the coauthors (40, 75). As blockade of estrogen synthesis with aromatase inhibitors or action with antiestrogens evolved, it became apparent that prevention of breast cancer with these agents might become possible. Trevor Powles pioneered the use of tamoxifen for breast cancer prevention (204). Many groups reported that high estradiol levels in postmenopausal women were associated with an increased risk of breast cancer (205, 206).

As responses to hormonal therapy were not uniform, a means was needed to predict which patients would respond to the antiestrogens and aromatase inhibitors. William McGuire (1937–1992), along with Elwood Jensen (1920–2012), had the vision to extensively correlate the presence of ERα with responsiveness of hormone-dependent breast cancer and to develop multiple concepts about breast cancer biology (62, 207–209). Use of selection for the presence of ERα allowed an enhancement of the percentage of responders to these agents. Interestingly, these studies demonstrated immediate and quite obvious efficacy of antiestrogens and aromatase inhibitors in a proportion of ER-positive patients (Supplemental Fig. 12) (26). The clear-cut responses observed support Charles Huggins’ statement that “if your therapy works, you do not need statistics to prove it” (the quotation is from Festschrift for Olaf Pearson, Case Western School of Medicine, 1987, as related personally to R.J.S.).

Recent attention has focused on resistance to antiestrogens and aromatase inhibitors in women with breast cancer. An early focus was on estrogen receptor mutations by Fuqua et al. (210–212) who described a K303R mutation that induced hypersensitivity to estradiol. An interesting mechanism is the emergence of clones of cells with mutations of the ER that render them capable of ligand-independent actions (108). These mutations are generally not present in tumors when initially discovered but are observed in tumors exposed long term to hormonal therapies (108), presumably through a process of selection. These observations have led to the more common use of the selective receptor downregulator fulvestrant and the development of new, more potent selective receptor downregulators (213, 214). The rationale is to eliminate these receptors as a means of abrogating their function.

Other syndromes of estrogen excess and deficiency

Benign tumors and malignant tumors containing high levels of estrogen can cause gynecomastia as in the Peutz-Jeghers syndrome and hepatocellular carcinoma (215–217). Duplication, deletion, and inversion resulting from subchromosomal recombinations result in syndromes of aromatase overexpression (218, 219). Inactivity of aromatase in women can result in pseudohermaphroditism, hirsutism, and other abnormalities (220–222).

Controversy

Many controversies arose with respect to the use of estrogen for treatment of the menopause, and both proponents and opponents expressed strong opinions (57, 87, 223–232). One controversy is whether estrogens cause or prevent breast cancer. The Women’s Health Initiative Trial suggests that conjugated equine estrogen, the estrogen used in that trial, reduced the risk of breast cancer by 23% (hazard ratio, 0.77; CI, 0.62 to 0.95) (233, 234) (Supplemental Fig. 13) (26). The mechanism for this is not clearly established but may reflect the proapoptotic effects of estrogens on occult, preexisting tumors (235–238). Interestingly, recent preclinical data suggest that the effects of conjugated equine estrogen may differ from those of estradiol (239). Another controversy is whether menopausal hormone therapy causes more benefit than harm in postmenopausal women. The recent Endocrine Society guideline concludes that benefits exceed harm in symptomatic women 50 to 59 years of age or <10 years postmenopausal when at low risk of breast cancer and coronary heart disease (231).

Complexity of estradiol effects

Recent reviews detail in considerable depth the actions of estrogen in women and in men and on brain (240), heart (114, 241), vasculature (114, 121), ovaries (125), bones (242), skeletal muscle (187), breast (205, 206, 243), adipose tissue (244), and reproductive tissue (104, 125). Studies of these actions in depth are found in the reviews cited above but were beyond the scope of the Clark Sawin Lecture.

Research into the various actions of estrogen has advanced rapidly in the past 5 years. To illustrate this point, the estrogen-related publications in the journal Endocrinology from 2013 until the present have been identified and categorized here. A major focus has been on the effects of estrogen on various aspects of brain function, including gonadotropin regulation, behavior, synaptic function, metabolism, and morphogenesis (245–280). Three other areas of emphasis have been bone (98, 281, 283), adrenal (284–287), and ovary (288–291). Other foci of study include pain mediation (292–294), uterus (295–297), glucose/insulin actions (298–302), and reproduction (303).

Topics and investigators left out

Any history must prioritize the issues raised, and this lecture illustrates this point well. We left out important effects on the brain, including behavior, gonadotropin regulation, and obsessive compulsive disorder, among others; cardiovascular actions; effects on the immune system; pubertal development; and management of menopause and actions on skeletal muscle. Finally, we apologize to the key investigators that we have failed to mention because of the time allotted to the lecture, but we refer readers to two reviews written by the coauthors (40, 75).

Abbreviations:

    Abbreviations:
     
  • DMBA

    dimethylbenzanthracene

  •  
  • ER

    estrogen receptor

Acknowledgments

Access to audio and video of Sawin lecture: https://figshare.com/articles/Sawinlecturemp4/7605488.

Disclosure Summary: The authors have nothing to disclose.

References

1.

Albright
F
,
Ellsberg
R
.
Uncharted Seas
.
Portland, OR
:
Kalmia Press
;
1990
:
3
95
.

2.

Paulshock
BZ
.
Tutankhamun and his brothers. Familial gynecomastia in the Eighteenth Dynasty
.
JAMA
.
1980
;
244
(
2
):
160
164
.

3.

Connell
SM
.
Aristotle and Galen on sex difference and reproduction: a new approach to an ancient rivalry
.
Stud Hist Philos Sci
.
2000
;
31
(
3
):
405
427
.

4.

Galanakis
E
.
Apgar score and Soranus of Ephesus
.
Lancet
.
1998
;
352
(
9145
):
2012
2013
.

5.

Karamanou
M
,
Tsoucalas
G
,
Creatsas
G
,
Androutsos
G
.
The effect of Soranus of Ephesus (98–138) on the work of midwives
.
Women Birth
.
2013
;
26
(
4
):
226
228
.

6.

Temkin
O
.
Soranus Gynecology
.
Baltimore, MD
:
Johns Hopkins University Press
;
1956
.

7.

Boylan
M
.
Galen
.
J Hist Biol
.
1986
;
19
(
1
):
47
77
.

8.

Hankinson
RJ
.
The Cambridge Companion to Galen
.
Cambridge, UK
:
Cambridge University Press
;
2009
.

9.

Kaplowitz
PB
.
Link between body fat and the timing of puberty. Pediatrics. 2008;121(Suppl 3):S208–S217
.

10.

Frisch
RE
,
Revelle
R
.
Height and weight at menarche and a hypothesis of menarche
.
Arch Dis Child
.
1971
;
46
(
249
):
695
701
.

11.

Frisch
RE
,
Revelle
R
.
The height and weight of girls and boys at the time of initiation of the adolescent growth spurt in height and weight and the relationship to menarche
.
Hum Biol
.
1971
;
43
(
1
):
140
159
.

12.

Frisch
RE
.
Body fat, menarche, fitness and fertility
.
Hum Reprod
.
1987
;
2
(
6
):
521
533
.

13.

Lorriaux
DL
.
Thomas Wharton (1614–1673): codifying the glands. In: A Biographical History of Endocrinology. Oxford, UK: John Wiley & Sons; 2016:49–54
.

14.

Mettler CC, Mettler FA. Gynecology in the nineteenth century: ovariotomy. In: History of Medicine. Philadelphia, PA: The Blakiston Company; 1947
.

15.

Gendrin AN. Chapter 13. In: Traité Philosophique de Médecine Pratique. Paris, France: Baillière; 1838:987–1003
.

16.

Bernard
C
.
An Introduction to the Study of Experimental Medicine. Reprinted in Classics of Medicine series from the original published in 1864 by Gazette Médicale de Paris. New York, NY: Macmillan Company; 1927
.

17.

Battey
R
.
Normal ovariotomectomy
.
Atlanta Medical and Surgical Journal
.
1873
;
10
:
321
339
.

18.

Knauer E. Ovarian transplantation in rabbits: a preliminary report. Contralb F Gyn., 1898
.

19.

Knauer E. On ovarian transplantation: labour and normal end of pregnancy after transplantation of ovaries in rabbits. Contralb F Gyn., no. 8, 1898
.

20.

Knauer E. Transplantation of the ovaries: an experimental study. Archiv F Gyn., 1898
.

21.

Fosbery
WH
.
Severe climacteric flushings successfully treated with ovarian extract
.
BMJ
.
1897
;
1
:
1039
.

22.

Brown-Séquard
C
.
Note on the effects produced on man by subcutaneous injections of a liquid obtained from the testicles of animals
.
Lancet
.
1889
;
134
(
3438
):
105
107
.

23.

Nieschlag
E
,
Nieschlag
S
.
Testosterone deficiency: a historical perspective
.
Asian J Androl
.
2014
;
16
(
2
):
161
168
.

24.

Easterbrook CC. Organo-therapeutics in mental diseases. BMJ. 1900;2:813–823
.

25.

Miao Sun Si. Essential Formulas for Emergencies [Worth] a Thousand Pieces of Gold [in Chinese]; 652
.

26.

Santen
RJ
,
Simpson
E
.
Data from: History of estrogen: its purification, structure, synthesis, biologic actions, and clinical implications. figshare 2019. Deposited 26 January 2019. https://figshare.com/articles/Supplemental figures History of Estrogen/7635968
.

27.

Temple
R
.
The science of endocrinology. In: The Genius of China: 3000 Years of Science, Discovery, and Invention. London, UK: Prion; 1998:127–130
.

28.

Beatson
GT
.
On the treatment of inoperable cases of carcinoma of the mamma: suggestions for a new method of treatment, with illustrative cases
.
Trans Med Chir Soc Edinb
.
1896
;
15
:
153
179
.

29.

Stockwell
S
.
Classics in oncology. George Thomas Beatson, M.D. (1848–1933)
.
CA Cancer J Clin
.
1983
;
33
(
2
):
105
121
.

30.

Clarke
MJ
.
Ovarian ablation in breast cancer, 1896 to 1998: milestones along hierarchy of evidence from case report to Cochrane review
.
BMJ
.
1998
;
317
(
7167
):
1246
1248
.

31.

Allen
E
,
Francis
BF
,
Robertson
LL
.
The hormone of the ovarian follicle: its localization and action in test animals and additional points bearing upon the internal secretion of the ovary
.
Am J Anat
.
1924
;
34
(
1
):
133
181
.

32.

Allen
E
,
Doisy
EA
.
An ovarian hormone: preliminary report on its localization, extraction and partial purification and action in test animal
.
JAMA
.
1923
;
81
(
10
):
819
821
.

33.

Doisy
EA
.
An autobiography
.
Annu Rev Biochem
.
1976
;
45
(
1
):
1
9
.

34.

Rohde
W
.
The contributions of Aschheim and Zondek to endocrinology
.
Pediatr Endocrinol Rev
.
2010
;
7
(
4
):
323
327
.

35.

Doisy
EA
,
Thayer
S
,
Veler
CD
.
The crystals of thefollicular ovarian hormone
.
Proc Soc Exp Biol Med
.
1930
;
27
(
5
):
417
419
.

36.

Veler
CD
,
Thayer
S
,
Doisy
EA
.
The preparation of the crystalline follicular ovarian hormone: theelin
.
J Biol Chem
.
1930
;
87
:
357
371
.

37.

Butenandt
A
.
Über “Progynonein” kystallisiertes weibliches Exualhormon
.
Naturwissenschaften
.
1929
;
17
(
45
):
879
.

38.

Huffman
MN
,
Thayer
SA
,
Doisy
EA
.
The isolation of alpha-dihydrotheelin from human placenta
.
J Biol Chem
.
1940
;
133
:
567
571
.

39.

Thayer
SA
,
Levin
L
,
Doisy
EA
.
Characterization of theelol
.
J Biol Chem
.
1931
;
91
:
655
665
.

40.

Simpson
E
,
Santen
RJ
.
Celebrating 75 years of oestradiol
.
J Mol Endocrinol
.
2015
;
55
(
3
):
T1
T20
.

41.

Albright
F
.
The effect of hormones on osteogenesis in man
.
Recent Prog Horm Res
.
1947
;
1
:
293
353
.

42.

Albright
F
,
Halsted
JA
,
Cloney
E
.
Studies on ovarian dysfunction
.
N Engl J Med
.
1935
;
212
(
5
):
192
195
.

43.

Albright
F
.
Studies on ovarian dysfunction. III. The menopause
.
Endocrinology
.
1936
;
20
:
24
39
.

44.

Forbes
AP
.
Fuller Albright. His concept of postmenopausal osteoporosis and what came of it. Clin Orthop Relat Res. 1991;(269):128–141
.

45.

Sturgis
SH
,
Albright
F
.
The mechanism of estrin therapy in the relief of dysmenorrhea
.
Endocrinology
.
1940
;
25
:
68
72
.

46.

Pfeiffer
CA
,
Gardner
WU
.
Skeletal changes and blood calcium level in pigeons receiving estrogens
.
Endocrinology
.
1938
;
23
:
485
491
.

47.

Reifenstein
EC
Jr,
Albright
F
.
The metabolic effects of steroid hormones in osteoporosis
.
J Clin Invest
.
1947
;
26
(
1
):
24
56
.

48.

Wallach
S
,
Henneman
PH
.
Prolonged estrogen therapy in postmenopausal women
.
J Am Med Assoc
.
1959
;
171
(
12
):
1637
1642
.

49.

Huggins
C
.
How Charles Huggins made his Nobel prize winning discovery—in his own words: an historic audio recording. Interviewed by Willard Goodwin and Elmer Bell
.
Prostate
.
2012
;
72
(
16
):
1718
.

50.

Huggins
C
,
Grand
LC
,
Brillantes
FP
.
Critical significance of breast structure in the induction of mammary cancer in the rat
.
Proc Natl Acad Sci USA
.
1959
;
45
(
8
):
1294
1300
.

51.

Huggins
C
,
Briziarelli
G
,
Sutton
H
Jr
.
Rapid induction of mammary carcinoma in the rat and the influence of hormones on the tumors
.
J Exp Med
.
1959
;
109
(
1
):
25
42
.

52.

Huggins
C
,
Grand
LC
,
Brillantes
FP
.
Mammary cancer induced by a single feeding of polynuclear hydrocarbons, and its suppression
.
Nature
.
1961
;
189
(
4760
):
204
207
.

53.

Huggins
C
,
Grand
L
,
Fukunishi
R
Aromatic influences on the yields of mammary cancers following administration of 7,12-dimethylbenz(a)anthracene
.
Proc Natl Acad Sci USA
.
1964
;
51
:
737
742
.

54.

Welsch
CW
.
Host factors affecting the growth of carcinogen-induced rat mammary carcinomas: a review and tribute to Charles Brenton Huggins
.
Cancer Res
.
1985
;
45
(
8
):
3415
3443
.

55.

Speroff
L
.
A Good Man: The Man, His Story, the Birth Control Pill
.
Portland, OR
:
Arnica Publishing
;
2009
.

56.

Goldzieher
JW
.
The history of steroidal contraceptive development: the estrogens
.
Perspect Biol Med
.
1993
;
36
(
3
):
363
368
.

57.

Watkins
ED
.
The Estrogen Elixir: A History of Hormone Replacement in America
.
Baltimore, MD
:
Johns Hopkins University Press
;
2007
.

58.

Colton
FB
.
Steroids and “the pill”: early steroid research at Searle
.
Steroids
.
1992
;
57
(
12
):
624
630
.

59.

Fehring
RJ
.
An analysis of the majority report “Responsible Parenthood” and its recommendations on abortion, sterilization, and contraception. In: Koterski JW, ed. Life and Learning XIII: Proceedings of the Thirteenth University Faculty for Life Conference: 2003 at Georgetown University. Washington, DC: University Faculty for Life; 2004: 120–152
.

60.

Goldin
C
,
Katz
LF
.
The power of the pill: oral contraceptives and women’s career and marriage decisions
.
J Polit Econ
.
2002
;
110
(
4
):
730
770
.

61.

Jensen
EV
.
On the mechanism of estrogen action
.
Perspect Biol Med
.
1962
;
6
(
1
):
47
59
.

62.

Block
GE
,
Ellis
RS
,
DeSombre
E
,
Jensen
E
.
Correlation of estrophilin content of primary mammary cancer to eventual endocrine treatment
.
Ann Surg
.
1978
;
188
(
3
):
372
376
.

63.

Zondek
B
.
Oesrogenic hormone in the urine of the stallion
.
Nature
.
1934
;
133
(
3361
):
494
.

64.

Steinach
E
,
Kun
H
.
Transformation of male sex hormones into a substance with the action of a female hormone
.
Lancet
.
1937
;
230
(
5954
):
845
.

65.

Balthazart
J
,
Ball
GF
.
New insights into the regulation and function of brain estrogen synthase (aromatase)
.
Trends Neurosci
.
1998
;
21
(
6
):
243
249
.

66.

Brodie
HJ
,
Pillai
AK
,
Hay
CE
.
Studies on the mechanisms of estrogen biosynthesis. VII. 2β-Hydroxylation of estr-4-ene-3,17-dione in human placenta, in vitro
.
Biochim Biophys Acta
.
1969
;
187
(
2
):
275
277
.

67.

Brodie
HJ
,
Raab
K
,
Possanza
G
,
Seto
N
,
Gut
M
.
Further stereochemical studies of the catalytic reduction of delta-1,4-3-keto steroids with tritium
.
J Org Chem
.
1969
;
34
(
9
):
2697
2702
.

68.

Brodie
HJ
,
Kripalani
KJ
,
Possanza
G
.
Studies on the mechanism of estrogen biosynthesis. VI. The stereochemistry of hydrogen elimination at C-2 during aromatization
.
J Am Chem Soc
.
1969
;
91
(
5
):
1241
1242
.

69.

Brodie
HJ
,
Possanza
G
,
Townsley
JD
.
Studies on the mechanism of estrogen biosynthesis. V. Stereochemical comparison of aromatization in placental and microbiological systems
.
Biochim Biophys Acta
.
1968
;
152
(
4
):
770
777
.

70.

Meyer
AS
.
19-Hydroxylation of Δ4-androstene-3,17-dione and dehydroepiandrosterone by bovine adrenals
.
Experientia
.
1955
;
11
(
3
):
99
102
.

71.

Meyer
AS
,
Hayano
M
,
Lindberg
MC
,
Gut
M
,
Rodgers
OG
.
The conversion of Δ4-androstene-3,17-dione-4-C14 and dehydroepiandrosterone by bovine adrenal homogenate preparations
.
Acta Endocrinol (Copenh)
.
1955
;
18
(
2
):
148
168
.

72.

Meyer
AS
.
Conversion of 19-hydroxy-Δ4-androstene-3,17-dione to estrone by endocrine tissue
.
Biochim Biophys Acta
.
1955
;
17
(
3
):
441
442
.

73.

Morato
T
,
Hayano
M
,
Dorfman
RI
,
Axelrod
LR
.
The intermediate steps in the biosynthesis of estrogens from androgens
.
Biochem Biophys Res Commun
.
1961
;
6
(
5
):
334
338
.

74.

Yoshimoto
FK
,
Guengerich
FP
.
Mechanism of the third oxidative step in the conversion of androgens to estrogens by cytochrome P450 19A1 steroid aromatase
.
J Am Chem Soc
.
2014
;
136
(
42
):
15016
15025
.

75.

Santen
RJ
,
Brodie
H
,
Simpson
ER
,
Siiteri
PK
,
Brodie
A
.
History of aromatase: saga of an important biological mediator and therapeutic target
.
Endocr Rev
.
2009
;
30
(
4
):
343
375
.

76.

Thompson
EA
Jr,
Siiteri
PK
.
The involvement of human placental microsomal cytochrome P-450 in aromatization
.
J Biol Chem
.
1974
;
249
(
17
):
5373
5378
.

77.

Thompson
EA
Jr,
Siiteri
PK
.
Utilization of oxygen and reduced nicotinamide adenine dinucleotide phosphate by human placental microsomes during aromatization of androstenedione
.
J Biol Chem
.
1974
;
249
(
17
):
5364
5372
.

78.

Thompson
EA
Jr,
Siiteri
PK
.
Subcellular distribution of aromatase in human placenta and ovary
.
Horm Res
.
1979
;
11
(
4
):
179
185
.

79.

Mendelson
CR
,
Wright
EE
,
Evans
CT
,
Porter
JC
,
Simpson
ER
.
Preparation and characterization of polyclonal and monoclonal antibodies against human aromatase cytochrome P-450 (P-450AROM), and their use in its purification
.
Arch Biochem Biophys
.
1985
;
243
(
2
):
480
491
.

80.

Pasanen
M
,
Pelkonen
O
.
Solubilization and partial purification of human placental cytochromes P-450
.
Biochem Biophys Res Commun
.
1981
;
103
(
4
):
1310
1317
.

81.

Osawa
Y
,
Yoshida
N
,
Fronckowiak
M
,
Kitawaki
J
.
Immunoaffinity purification of aromatase cytochrome P-450 from human placental microsomes, metabolic switching from aromatization to 1β and 2β-monohydroxylation, and recognition of aromatase isozymes
.
Steroids
.
1987
;
50
(
1–3
):
11
28
.

82.

Hall
PF
,
Chen
S
,
Nakajin
S
,
Shinoda
M
,
Shively
JE
.
Purification and characterization of aromatase from human placenta
.
Steroids
.
1987
;
50
(
1–3
):
37
50
.

83.

Muto
N
,
Tan
L
.
Purification of oestrogen synthetase by high-performance liquid chromatography. Two membrane-bound enzymes from the human placenta
.
J Chromatogr A
.
1985
;
326
:
137
146
.

84.

Kellis
JT
Jr,
Vickery
LE
.
Purification and characterization of human placental aromatase cytochrome P-450
.
J Biol Chem
.
1987
;
262
(
9
):
4413
4420
.

85.

Corbin
CJ
,
Graham-Lorence
S
,
McPhaul
M
,
Mason
JI
,
Mendelson
CR
,
Simpson
ER
.
Isolation of a full-length cDNA insert encoding human aromatase system cytochrome P-450 and its expression in nonsteroidogenic cells
.
Proc Natl Acad Sci USA
.
1988
;
85
(
23
):
8948
8952
.

86.

Harada
N
.
Cloning of a complete cDNA encoding human aromatase: immunochemical identification and sequence analysis
.
Biochem Biophys Res Commun
.
1988
;
156
(
2
):
725
732
.

87.

McPhaul
MJ
,
Noble
JF
,
Matsumine
H
,
Wilson
JD
.
Cloning and expression of the chicken ovary aromatase P-450: expression of mRNA in tissues of the Sebright and Leghorn chicken
.
Trans Assoc Am Physicians
.
1988
;
101
:
219
225
.

88.

McPhaul
MJ
,
Noble
JF
,
Simpson
ER
,
Mendelson
CR
,
Wilson
JD
.
The expression of a functional cDNA encoding the chicken cytochrome P-450arom (aromatase) that catalyzes the formation of estrogen from androgen
.
J Biol Chem
.
1988
;
263
(
31
):
16358
16363
.

89.

Toda
K
,
Terashima
M
,
Mitsuuchi
Y
,
Yamasaki
Y
,
Yokoyama
Y
,
Nojima
S
,
Ushiro
H
,
Maeda
T
,
Yamamoto
Y
,
Sagara
Y
,
Shizuta
Y
.
Alternative usage of different poly(A) addition signals for two major species of mRNA encoding human aromatase P-450
.
FEBS Lett
.
1989
;
247
(
2
):
371
376
.

90.

Hickey
GJ
,
Krasnow
JS
,
Beattie
WG
,
Richards
JS
.
Aromatase cytochrome P450 in rat ovarian granulosa cells before and after luteinization: adenosine 3′,5′-monophosphate-dependent and independent regulation. Cloning and sequencing of rat aromatase cDNA and 5′ genomic DNA
.
Mol Endocrinol
.
1990
;
4
(
1
):
3
12
.

91.

Terashima
M
,
Toda
K
,
Kawamoto
T
,
Kuribayashi
I
,
Ogawa
Y
,
Maeda
T
,
Shizuta
Y
.
Isolation of a full-length cDNA encoding mouse aromatase P450
.
Arch Biochem Biophys
.
1991
;
285
(
2
):
231
237
.

92.

Tanaka
M
,
Telecky
TM
,
Fukada
S
,
Adachi
S
,
Chen
S
,
Nagahama
Y
.
Cloning and sequence analysis of the cDNA encoding P-450 aromatase (P450arom) from a rainbow trout (Oncorhynchus mykiss) ovary; relationship between the amount of P450arom mRNA and the production of oestradiol-17β in the ovary
.
J Mol Endocrinol
.
1992
;
8
(
1
):
53
61
.

93.

Laughton
CA
,
Zvelebil
MJ
,
Neidle
S
.
A detailed molecular model for human aromatase
.
J Steroid Biochem Mol Biol
.
1993
;
44
(
4–6
):
399
407
.

94.

Graham-Lorence
S
,
Amarneh
B
,
White
RE
,
Peterson
JA
,
Simpson
ER
.
A three-dimensional model of aromatase cytochrome P450
.
Protein Sci
.
1995
;
4
(
6
):
1065
1080
.

95.

Williams
PA
,
Cosme
J
,
Sridhar
V
,
Johnson
EF
,
McRee
DE
.
Mammalian microsomal cytochrome P450 monooxygenase: structural adaptations for membrane binding and functional diversity
.
Mol Cell
.
2000
;
5
(
1
):
121
131
.

96.

Favia
AD
,
Cavalli
A
,
Masetti
M
,
Carotti
A
,
Recanatini
M
.
Three-dimensional model of the human aromatase enzyme and density functional parameterization of the iron-containing protoporphyrin IX for a molecular dynamics study of heme-cysteinato cytochromes
.
Proteins
.
2006
;
62
(
4
):
1074
1087
.

97.

Koymans
LM
,
Moereels
H
,
Vanden Bossche
H
.
A molecular model for the interaction between vorozole and other non-steroidal inhibitors and human cytochrome P450 19 (P450 aromatase)
.
J Steroid Biochem Mol Biol
.
1995
;
53
(
1–6
):
191
197
.

98.

Chen
S
,
Zhang
F
,
Sherman
MA
,
Kijima
I
,
Cho
M
,
Yuan
YC
,
Toma
Y
,
Osawa
Y
,
Zhou
D
,
Eng
ET
.
Structure–function studies of aromatase and its inhibitors: a progress report
.
J Steroid Biochem Mol Biol
.
2003
;
86
(
3–5
):
231
237
.

99.

Ghosh
D
,
Griswold
J
,
Erman
M
,
Pangborn
W
.
Structural basis for androgen specificity and oestrogen synthesis in human aromatase
.
Nature
.
2009
;
457
(
7226
):
219
223
.

100.

Harada
N
,
Utsumi
T
,
Takagi
Y
.
Tissue-specific expression of the human aromatase cytochrome P-450 gene by alternative use of multiple exons 1 and promoters, and switching of tissue-specific exons 1 in carcinogenesis
.
Proc Natl Acad Sci USA
.
1993
;
90
(
23
):
11312
11316
.

101.

Mahendroo
MS
,
Means
GD
,
Mendelson
CR
,
Simpson
ER
.
Tissue-specific expression of human P-450AROM. The promoter responsible for expression in adipose tissue is different from that utilized in placenta
.
J Biol Chem
.
1991
;
266
(
17
):
11276
11281
.

102.

Means
GD
,
Kilgore
MW
,
Mahendroo
MS
,
Mendelson
CR
,
Simpson
ER
.
Tissue-specific promoters regulate aromatase cytochrome P450 gene expression in human ovary and fetal tissues
.
Mol Endocrinol
.
1991
;
5
(
12
):
2005
2013
.

103.

Bulun
SE
,
Takayama
K
,
Suzuki
T
,
Sasano
H
,
Yilmaz
B
,
Sebastian
S
.
Organization of the human aromatase p450 (CYP19) gene
.
Semin Reprod Med
.
2004
;
22
(
1
):
5
9
.

104.

Hamilton
KJ
,
Hewitt
SC
,
Arao
Y
,
Korach
KS
.
Estrogen hormone biology
.
Curr Top Dev Biol
.
2017
;
125
:
109
146
.

105.

Mangelsdorf
DJ
,
Thummel
C
,
Beato
M
,
Herrlich
P
,
Schütz
G
,
Umesono
K
,
Blumberg
B
,
Kastner
P
,
Mark
M
,
Chambon
P
,
Evans
RM
.
The nuclear receptor superfamily: the second decade
.
Cell
.
1995
;
83
(
6
):
835
839
.

106.

Hewitt
SC
,
Korach
KS
.
Estrogen receptors: new directions in the new millennium
.
Endocr Rev
.
2018
;
39
(
5
):
664
675
.

107.

Aagaard
MM
,
Siersbæk
R
,
Mandrup
S
.
Molecular basis for gene-specific transactivation by nuclear receptors
.
Biochim Biophys Acta
.
2011
;
1812
(
8
):
824
835
.

108.

Jeselsohn
R
,
Bergholz
JS
,
Pun
M
,
Cornwell
M
,
Liu
W
,
Nardone
A
,
Xiao
T
,
Li
W
,
Qiu
X
,
Buchwalter
G
,
Feiglin
A
,
Abell-Hart
K
,
Fei
T
,
Rao
P
,
Long
H
,
Kwiatkowski
N
,
Zhang
T
,
Gray
N
,
Melchers
D
,
Houtman
R
,
Liu
XS
,
Cohen
O
,
Wagle
N
,
Winer
EP
,
Zhao
J
,
Brown
M
.
Allele-specific chromatin recruitment and therapeutic vulnerabilities of ESR1 activating mutations
.
Cancer Cell
.
2018
;
33
(
2
):
173
186.e5
.

109.

Couse
JF
,
Lindzey
J
,
Grandien
K
,
Gustafsson
JA
,
Korach
KS
.
Tissue distribution and quantitative analysis of estrogen receptor-α (ERα) and estrogen receptor-β (ERβ) messenger ribonucleic acid in the wild-type and ERα-knockout mouse
.
Endocrinology
.
1997
;
138
(
11
):
4613
4621
.

110.

Binder
AK
,
Rodriguez
KF
,
Hamilton
KJ
,
Stockton
PS
,
Reed
CE
,
Korach
KS
.
The absence of ER-β results in altered gene expression in ovarian granulosa cells isolated from in vivo preovulatory follicles
.
Endocrinology
.
2013
;
154
(
6
):
2174
2187
.

111.

Yager
JD
,
Chen
JQ
.
Mitochondrial estrogen receptors—new insights into specific functions
.
Trends Endocrinol Metab
.
2007
;
18
(
3
):
89
91
.

112.

Levin
ER
.
Extranuclear steroid receptors are essential for steroid hormone actions
.
Annu Rev Med
.
2015
;
66
(
1
):
271
280
.

113.

Klinge
CM
.
Estrogens regulate life and death in mitochondria
.
J Bioenerg Biomembr
.
2017
;
49
(
4
):
307
324
.

114.

Kim
JK
,
Levin
ER
.
Estrogen signaling in the cardiovascular system. Nucl Recept Signal. 2006;4:e013
.

115.

Madak-Erdogan
Z
,
Kieser
KJ
,
Kim
SH
,
Komm
B
,
Katzenellenbogen
JA
,
Katzenellenbogen
BS
.
Nuclear and extranuclear pathway inputs in the regulation of global gene expression by estrogen receptors
.
Mol Endocrinol
.
2008
;
22
(
9
):
2116
2127
.

116.

Levin
ER
.
Hormone action and breast cancer
.
Cancer Treat Res
.
2009
;
147
:
1
10
.

117.

Levin
ER
.
Extranuclear estrogen receptor’s roles in physiology: lessons from mouse models
.
Am J Physiol Endocrinol Metab
.
2014
;
307
(
2
):
E133
E140
.

118.

Levin
ER
.
Translating extranuclear steroid receptor signaling to clinical medicine
.
Horm Cancer
.
2014
;
5
(
3
):
140
145
.

119.

Levin
ER
,
Hammes
SR
.
Nuclear receptors outside the nucleus: extranuclear signalling by steroid receptors
.
Nat Rev Mol Cell Biol
.
2016
;
17
(
12
):
783
797
.

120.

Levin
ER
.
Membrane estrogen receptors signal to determine transcription factor function
.
Steroids
.
2018
;
132
:
1
4
.

121.

Chambliss
KL
,
Wu
Q
,
Oltmann
S
,
Konaniah
ES
,
Umetani
M
,
Korach
KS
,
Thomas
GD
,
Mineo
C
,
Yuhanna
IS
,
Kim
SH
,
Madak-Erdogan
Z
,
Maggi
A
,
Dineen
SP
,
Roland
CL
,
Hui
DY
,
Brekken
RA
,
Katzenellenbogen
JA
,
Katzenellenbogen
BS
,
Shaul
PW
.
Non-nuclear estrogen receptor alpha signaling promotes cardiovascular protection but not uterine or breast cancer growth in mice
.
J Clin Invest
.
2010
;
120
(
7
):
2319
2330
.

122.

Schultz-Norton
JR
,
Ziegler
YS
,
Nardulli
AM
.
ERα-associated protein networks
.
Trends Endocrinol Metab
.
2011
;
22
(
4
):
124
129
.

123.

Filardo
EJ
,
Thomas
P
.
Minireview: G protein-coupled estrogen receptor-1, GPER-1: its mechanism of action and role in female reproductive cancer, renal and vascular physiology
.
Endocrinology
.
2012
;
153
(
7
):
2953
2962
.

124.

Arnal
JF
,
Fontaine
C
,
Abot
A
,
Valera
MC
,
Laurell
H
,
Gourdy
P
,
Lenfant
F
.
Lessons from the dissection of the activation functions (AF-1 and AF-2) of the estrogen receptor alpha in vivo
.
Steroids
.
2013
;
78
(
6
):
576
582
.

125.

Hamilton
KJ
,
Arao
Y
,
Korach
KS
.
Estrogen hormone physiology: reproductive findings from estrogen receptor mutant mice
.
Reprod Biol
.
2014
;
14
(
1
):
3
8
.

126.

Magnani
L
,
Lupien
M
.
Chromatin and epigenetic determinants of estrogen receptor alpha (ESR1) signaling
.
Mol Cell Endocrinol
.
2014
;
382
(
1
):
633
641
.

127.

Kuiper
GG
,
Enmark
E
,
Pelto-Huikko
M
,
Nilsson
S
,
Gustafsson
JA
.
Cloning of a novel receptor expressed in rat prostate and ovary
.
Proc Natl Acad Sci USA
.
1996
;
93
(
12
):
5925
5930
.

128.

Hewitt
SC
,
Li
L
,
Grimm
SA
,
Chen
Y
,
Liu
L
,
Li
Y
,
Bushel
PR
,
Fargo
D
,
Korach
KS
.
Research resource: whole-genome estrogen receptor α binding in mouse uterine tissue revealed by ChIP-seq
.
Mol Endocrinol
.
2012
;
26
(
5
):
887
898
.

129.

Carroll
JS
,
Liu
XS
,
Brodsky
AS
,
Li
W
,
Meyer
CA
,
Szary
AJ
,
Eeckhoute
J
,
Shao
W
,
Hestermann
EV
,
Geistlinger
TR
,
Fox
EA
,
Silver
PA
,
Brown
M
.
Chromosome-wide mapping of estrogen receptor binding reveals long-range regulation requiring the forkhead protein FoxA1
.
Cell
.
2005
;
122
(
1
):
33
43
.

130.

Walter
P
,
Green
S
,
Greene
G
,
Krust
A
,
Bornert
JM
,
Jeltsch
JM
,
Staub
A
,
Jensen
E
,
Scrace
G
,
Waterfield
M
.
Cloning of the human estrogen receptor cDNA
.
Proc Natl Acad Sci USA
.
1985
;
82
(
23
):
7889
7893
.

131.

Fanning
SW
,
Mayne
CG
,
Dharmarajan
V
,
Carlson
KE
,
Martin
TA
,
Novick
SJ
,
Toy
W
,
Green
B
,
Panchamukhi
S
,
Katzenellenbogen
BS
,
Tajkhorshid
E
,
Griffin
PR
,
Shen
Y
,
Chandarlapaty
S
,
Katzenellenbogen
JA
,
Greene
GL
.
Estrogen receptor alpha somatic mutations Y537S and D538G confer breast cancer endocrine resistance by stabilizing the activating function-2 binding conformation
.
eLife
.
2016
;
5
:
e12792
.

132.

Singhal
H
,
Greene
ME
,
Zarnke
AL
,
Laine
M
,
Al Abosy
R
,
Chang
YF
,
Dembo
AG
,
Schoenfelt
K
,
Vadhi
R
,
Qiu
X
,
Rao
P
,
Santhamma
B
,
Nair
HB
,
Nickisch
KJ
,
Long
HW
,
Becker
L
,
Brown
M
,
Greene
GL
.
Progesterone receptor isoforms, agonists and antagonists differentially reprogram estrogen signaling
.
Oncotarget
.
2017
;
9
(
4
):
4282
4300
.

133.

Singhal
H
,
Greene
ME
,
Tarulli
G
,
Zarnke
AL
,
Bourgo
RJ
,
Laine
M
,
Chang
YF
,
Ma
S
,
Dembo
AG
,
Raj
GV
,
Hickey
TE
,
Tilley
WD
,
Greene
GL
.
Genomic agonism and phenotypic antagonism between estrogen and progesterone receptors in breast cancer
.
Sci Adv
.
2016
;
2
(
6
):
e1501924
.

134.

Börjesson
AE
,
Farman
HH
,
Movérare-Skrtic
S
,
Engdahl
C
,
Antal
MC
,
Koskela
A
,
Tuukkanen
J
,
Carlsten
H
,
Krust
A
,
Chambon
P
,
Sjögren
K
,
Lagerquist
MK
,
Windahl
SH
,
Ohlsson
C
.
SERMs have substance-specific effects on bone, and these effects are mediated via ERαAF-1 in female mice
.
Am J Physiol Endocrinol Metab
.
2016
;
310
(
11
):
E912
E918
.

135.

Murakami
G
,
Hojo
Y
,
Ogiue-Ikeda
M
,
Mukai
H
,
Chambon
P
,
Nakajima
K
,
Ooishi
Y
,
Kimoto
T
,
Kawato
S
.
Estrogen receptor KO mice study on rapid modulation of spines and long-term depression in the hippocampus
.
Brain Res
.
2015
;
1621
:
133
146
.

136.

Dahlman-Wright
K
,
Cavailles
V
,
Fuqua
SA
,
Jordan
VC
,
Katzenellenbogen
JA
,
Korach
KS
,
Maggi
A
,
Muramatsu
M
,
Parker
MG
,
Gustafsson
JA
.
International Union of Pharmacology. LXIV. Estrogen receptors
.
Pharmacol Rev
.
2006
;
58
(
4
):
773
781
.

137.

Nilsson
S
,
Mäkelä
S
,
Treuter
E
,
Tujague
M
,
Thomsen
J
,
Andersson
G
,
Enmark
E
,
Pettersson
K
,
Warner
M
,
Gustafsson
JA
.
Mechanisms of estrogen action
.
Physiol Rev
.
2001
;
81
(
4
):
1535
1565
.

138.

Rajapaksa
G
,
Thomas
C
,
Gustafsson
JA
.
Estrogen signaling and unfolded protein response in breast cancer
.
J Steroid Biochem Mol Biol
.
2016
;
163
:
45
50
.

139.

Thomas
C
,
Gustafsson
JA
.
Estrogen receptor mutations and functional consequences for breast cancer
.
Trends Endocrinol Metab
.
2015
;
26
(
9
):
467
476
.

140.

Warner
M
,
Huang
B
,
Gustafsson
JA
.
Estrogen receptor β as a pharmaceutical target
.
Trends Pharmacol Sci
.
2017
;
38
(
1
):
92
99
.

141.

Chute
JP
,
Ross
JR
,
McDonnell
DP
.
Minireview: nuclear receptors, hematopoiesis, and stem cells
.
Mol Endocrinol
.
2010
;
24
(
1
):
1
10
.

142.

Nelson
ER
,
Wardell
SE
,
McDonnell
DP
.
The molecular mechanisms underlying the pharmacological actions of estrogens, SERMs and oxysterols: implications for the treatment and prevention of osteoporosis
.
Bone
.
2013
;
53
(
1
):
42
50
.

143.

Gorski
J
,
Wendell
D
,
Gregg
D
,
Chun
TY
.
Estrogens and the genetic control of tumor growth
.
Prog Clin Biol Res
.
1997
;
396
:
233
243
.

144.

Gorski
J
,
Hou
Q.
Embryonic estrogen receptors: do they have a physiological function? Environ Health Perspect. 1995;103 (Suppl 7):69–72
.

145.

Watters
JJ
,
Chun
TY
,
Kim
YN
,
Bertics
PJ
,
Gorski
J
.
Estrogen modulation of prolactin gene expression requires an intact mitogen-activated protein kinase signal transduction pathway in cultured rat pituitary cells
.
Mol Endocrinol
.
2000
;
14
(
11
):
1872
1881
.

146.

Hewitt
SC
,
Winuthayanon
W
,
Korach
KS
.
What’s new in estrogen receptor action in the female reproductive tract
.
J Mol Endocrinol
.
2016
;
56
(
2
):
R55
R71
.

147.

Smith
EP
,
Boyd
J
,
Frank
GR
,
Takahashi
H
,
Cohen
RM
,
Specker
B
,
Williams
TC
,
Lubahn
DB
,
Korach
KS
.
Estrogen resistance caused by a mutation in the estrogen-receptor gene in a man
.
N Engl J Med
.
1994
;
331
(
16
):
1056
1061
.

148.

Bulun
SE
,
Fang
Z
,
Imir
G
,
Gurates
B
,
Tamura
M
,
Yilmaz
B
,
Langoi
D
,
Amin
S
,
Yang
S
,
Deb
S
.
Aromatase and endometriosis
.
Semin Reprod Med
.
2004
;
22
(
1
):
45
50
.

149.

Bulun
SE
,
Monsivais
D
,
Kakinuma
T
,
Furukawa
Y
,
Bernardi
L
,
Pavone
ME
,
Dyson
M
.
Molecular biology of endometriosis: from aromatase to genomic abnormalities
.
Semin Reprod Med
.
2015
;
33
(
3
):
220
224
.

150.

Pavone
ME
,
Bulun
SE
.
Clinical review: the use of aromatase inhibitors for ovulation induction and superovulation
.
J Clin Endocrinol Metab
.
2013
;
98
(
5
):
1838
1844
.

151.

Zhao
H
,
Zhou
L
,
Shangguan
AJ
,
Bulun
SE
.
Aromatase expression and regulation in breast and endometrial cancer
.
J Mol Endocrinol
.
2016
;
57
(
1
):
R19
R33
.

152.

Frasor
J
,
Barnett
DH
,
Danes
JM
,
Hess
R
,
Parlow
AF
,
Katzenellenbogen
BS
.
Response-specific and ligand dose-dependent modulation of estrogen receptor (ER) α activity by ERβ in the uterus
.
Endocrinology
.
2003
;
144
(
7
):
3159
3166
.

153.

Harrington
WR
,
Kim
SH
,
Funk
CC
,
Madak-Erdogan
Z
,
Schiff
R
,
Katzenellenbogen
JA
,
Katzenellenbogen
BS
.
Estrogen dendrimer conjugates that preferentially activate extranuclear, nongenomic versus genomic pathways of estrogen action
.
Mol Endocrinol
.
2006
;
20
(
3
):
491
502
.

154.

Montano
MM
,
Bianco
NR
,
Deng
H
,
Wittmann
BM
,
Chaplin
LC
,
Katzenellenbogen
BS
.
Estrogen receptor regulation of quinone reductase in breast cancer: implications for estrogen-induced breast tumor growth and therapeutic uses of tamoxifen
.
Front Biosci
.
2005
;
10
(
1–3
):
1440
1461
.

155.

Tryfonidis
K
,
Zardavas
D
,
Katzenellenbogen
BS
,
Piccart
M
.
Endocrine treatment in breast cancer: cure, resistance and beyond
.
Cancer Treat Rev
.
2016
;
50
:
68
81
.

156.

Dasgupta
S
,
O’Malley
BW
.
Transcriptional coregulators: emerging roles of SRC family of coactivators in disease pathology
.
J Mol Endocrinol
.
2014
;
53
(
2
):
R47
R59
.

157.

Foulds
CE
,
Panigrahi
AK
,
Coarfa
C
,
Lanz
RB
,
O’Malley
BW
.
Long noncoding RNAs as targets and regulators of nuclear receptors
.
Curr Top Microbiol Immunol
.
2016
;
394
:
143
176
.

158.

McKenna
NJ
,
Evans
RM
,
O’Malley
BW
.
Nuclear receptor signaling: a home for nuclear receptor and coregulator signaling research. Nucl Recept Signal. 2014;12:e006.

159.

Power
RF
,
Conneely
OM
,
O’Malley
BW
.
New insights into activation of the steroid hormone receptor superfamily
.
Trends Pharmacol Sci
.
1992
;
13
(
8
):
318
323
.

160.

Szwarc
MM
,
Lydon
JP
,
O’Malley
BW
.
Steroid receptor coactivators as therapeutic targets in the female reproductive system
.
J Steroid Biochem Mol Biol
.
2015
;
154
:
32
38
.

161.

Wang
L
,
Lonard
DM
,
O’Malley
BW
.
The role of steroid receptor coactivators in hormone dependent cancers and their potential as therapeutic targets
.
Horm Cancer
.
2016
;
7
(
4
):
229
235
.

162.

Pietras
RJ
,
Szego
CM
.
Specific binding sites for oestrogen at the outer surfaces of isolated endometrial cells
.
Nature
.
1977
;
265
(
5589
):
69
72
.

163.

Yang
SH
,
Liu
R
,
Perez
EJ
,
Wen
Y
,
Stevens
SM
Jr
,
Valencia
T
,
Brun-Zinkernagel
AM
,
Prokai
L
,
Will
Y
,
Dykens
J
,
Koulen
P
,
Simpkins
JW
.
Mitochondrial localization of estrogen receptor β
.
Proc Natl Acad Sci USA
.
2004
;
101
(
12
):
4130
4135
.

164.

Song
RX
.
Membrane-initiated steroid signaling action of estrogen and breast cancer
.
Semin Reprod Med
.
2007
;
25
(
3
):
187
197
.

165.

Song
RX
,
Chen
Y
,
Zhang
Z
,
Bao
Y
,
Yue
W
,
Wang
JP
,
Fan
P
,
Santen
RJ
.
Estrogen utilization of IGF-1-R and EGF-R to signal in breast cancer cells
.
J Steroid Biochem Mol Biol
.
2010
;
118
(
4-5
):
219
230
.

166.

Haas
E
,
Bhattacharya
I
,
Brailoiu
E
,
Damjanović
M
,
Brailoiu
GC
,
Gao
X
,
Mueller-Guerre
L
,
Marjon
NA
,
Gut
A
,
Minotti
R
,
Meyer
MR
,
Amann
K
,
Ammann
E
,
Perez-Dominguez
A
,
Genoni
M
,
Clegg
DJ
,
Dun
NJ
,
Resta
TC
,
Prossnitz
ER
,
Barton
M
.
Regulatory role of G protein–coupled estrogen receptor for vascular function and obesity
.
Circ Res
.
2009
;
104
(
3
):
288
291
.

167.

Pedram
A
,
Razandi
M
,
Kim
JK
,
O’Mahony
F
,
Lee
EY
,
Luderer
U
,
Levin
ER
.
Developmental phenotype of a membrane only estrogen receptor α (MOER) mouse
.
J Biol Chem
.
2009
;
284
(
6
):
3488
3495
.

168.

Li
Y
,
Wang
JP
,
Santen
RJ
,
Kim
TH
,
Park
H
,
Fan
P
,
Yue
W
.
Estrogen stimulation of cell migration involves multiple signaling pathway interactions
.
Endocrinology
.
2010
;
151
(
11
):
5146
5156
.

169.

Flamini
MI
,
Sanchez
AM
,
Genazzani
AR
,
Simoncini
T
.
Estrogen regulates endometrial cell cytoskeletal remodeling and motility via focal adhesion kinase
.
Fertil Steril
.
2011
;
95
(
2
):
722
726
.

170.

Sanchez
AM
,
Flamini
MI
,
Zullino
S
,
Gopal
S
,
Genazzani
AR
,
Simoncini
T
.
Estrogen receptor-α promotes endothelial cell motility through focal adhesion kinase
.
Mol Hum Reprod
.
2011
;
17
(
4
):
219
226
.

171.

Wu
Q
,
Chambliss
K
,
Umetani
M
,
Mineo
C
,
Shaul
PW
.
Non-nuclear estrogen receptor signaling in the endothelium
.
J Biol Chem
.
2011
;
286
(
17
):
14737
14743
.

172.

Razandi
M
,
Pedram
A
,
Jordan
VC
,
Fuqua
S
,
Levin
ER
.
Tamoxifen regulates cell fate through mitochondrial estrogen receptor beta in breast cancer
.
Oncogene
.
2013
;
32
(
27
):
3274
3285
.

173.

Marjon
NA
,
Hu
C
,
Hathaway
HJ
,
Prossnitz
ER
.
G protein–coupled estrogen receptor regulates mammary tumorigenesis and metastasis
.
Mol Cancer Res
.
2014
;
12
(
11
):
1644
1654
.

174.

Razandi
M
,
Alton
G
,
Pedram
A
,
Ghonshani
S
,
Webb
P
,
Levin
ER
.
Identification of a structural determinant necessary for the localization and function of estrogen receptor α at the plasma membrane
.
Mol Cell Biol
.
2003
;
23
(
5
):
1633
1646
.

175.

Song
RX
,
Barnes
CJ
,
Zhang
Z
,
Bao
Y
,
Kumar
R
,
Santen
RJ
.
The role of Shc and insulin-like growth factor 1 receptor in mediating the translocation of estrogen receptor α to the plasma membrane
.
Proc Natl Acad Sci USA
.
2004
;
101
(
7
):
2076
2081
.

176.

Song
RX
,
Zhang
Z
,
Chen
Y
,
Bao
Y
,
Santen
RJ
.
Estrogen signaling via a linear pathway involving insulin-like growth factor I receptor, matrix metalloproteinases, and epidermal growth factor receptor to activate mitogen-activated protein kinase in MCF-7 breast cancer cells
.
Endocrinology
.
2007
;
148
(
8
):
4091
4101
.

177.

Madak-Erdogan
Z
,
Lupien
M
,
Stossi
F
,
Brown
M
,
Katzenellenbogen
BS
.
Genomic collaboration of estrogen receptor α and extracellular signal-regulated kinase 2 in regulating gene and proliferation programs
.
Mol Cell Biol
.
2011
;
31
(
1
):
226
236
.

178.

Madak-Erdogan
Z
,
Charn
TH
,
Jiang
Y
,
Liu
ET
,
Katzenellenbogen
JA
,
Katzenellenbogen
BS
.
Integrative genomics of gene and metabolic regulation by estrogen receptors α and β, and their coregulators
.
Mol Syst Biol
.
2017
;
13
(
4
):
929
.

179.

Zheng
FF
,
Wu
RC
,
Smith
CL
,
O’Malley
BW
.
Rapid estrogen-induced phosphorylation of the SRC-3 coactivator occurs in an extranuclear complex containing estrogen receptor
.
Mol Cell Biol
.
2005
;
25
(
18
):
8273
8284
.

180.

Maffei
L
,
Murata
Y
,
Rochira
V
,
Tubert
G
,
Aranda
C
,
Vazquez
M
,
Clyne
CD
,
Davis
S
,
Simpson
ER
,
Carani
C
.
Dysmetabolic syndrome in a man with a novel mutation of the aromatase gene: effects of testosterone, alendronate, and estradiol treatment
.
J Clin Endocrinol Metab
.
2004
;
89
(
1
):
61
70
.

181.

Robertson
KM
,
O’Donnell
L
,
Jones
ME
,
Meachem
SJ
,
Boon
WC
,
Fisher
CR
,
Graves
KH
,
McLachlan
RI
,
Simpson
ER
.
Impairment of spermatogenesis in mice lacking a functional aromatase (cyp 19) gene
.
Proc Natl Acad Sci USA
.
1999
;
96
(
14
):
7986
7991
.

182.

Jones
ME
,
Boon
WC
,
Proietto
J
,
Simpson
ER
.
Of mice and men: the evolving phenotype of aromatase deficiency
.
Trends Endocrinol Metab
.
2006
;
17
(
2
):
55
64
.

183.

Simpson
ER
.
Models of aromatase insufficiency
.
Semin Reprod Med
.
2004
;
22
(
1
):
25
30
.

184.

Kirma
N
,
Gill
K
,
Mandava
U
,
Tekmal
RR
.
Overexpression of aromatase leads to hyperplasia and changes in the expression of genes involved in apoptosis, cell cycle, growth, and tumor suppressor functions in the mammary glands of transgenic mice
.
Cancer Res
.
2001
;
61
(
5
):
1910
1918
.

185.

Mandava
U
,
Kirma
N
,
Tekmal
RR
.
Aromatase overexpression transgenic mice model: cell type specific expression and use of letrozole to abrogate mammary hyperplasia without affecting normal physiology
.
J Steroid Biochem Mol Biol
.
2001
;
79
(
1–5
):
27
34
.

186.

Li
X
,
Strauss
L
,
Mäkelä
S
,
Streng
T
,
Huhtaniemi
I
,
Santti
R
,
Poutanen
M
.
Multiple structural and functional abnormalities in the p450 aromatase expressing transgenic male mice are ameliorated by a p450 aromatase inhibitor
.
Am J Pathol
.
2004
;
164
(
3
):
1039
1048
.

187.

Finkelstein
JS
,
Yu
EW
,
Burnett-Bowie
SA
.
Gonadal steroids and body composition, strength, and sexual function in men
.
N Engl J Med
.
2013
;
369
(
25
):
2457
.

188.

Siiteri PK, MacDonald PC. Role of extraglandular estrogen in human endocrinology. In: Greep RO, Astwood E, eds. Handbook of Physiology: Endocrinology. Vol 2, Pt 1. Washington, DC, American Physiological Society; 1973;615–629
.

189.

Huggins
C
.
Endocrine substances in the treatment of cancers
.
J Am Med Assoc
.
1949
;
141
(
11
):
750
754
.

190.

Talalay
P
,
Takano
GM
,
Huggins
C
.
Studies on the Walker tumor. II. Effects of adrenalectomy and hypophysectomy on tumor growth in tube-fed rats
.
Cancer Res
.
1952
;
12
(
11
):
838
843
.

191.

Herbst
AL
,
Griffiths
CT
,
Kistner
RW
.
Clomiphene citrate (NSC-35770) in disseminated mammary carcinoma
.
Cancer Chemother Rep
.
1964
;
43
:
39
41
.

192.

Cole
MP
,
Jones
CT
,
Todd
ID
.
A new anti-oestrogenic agent in late breast cancer. An early clinical appraisal of ICI46474
.
Br J Cancer
.
1971
;
25
(
2
):
270
275
.

193.

Ward
HW
.
Anti-oestrogen therapy for breast cancer: a trial of tamoxifen at two dose levels
.
BMJ
.
1973
;
1
(
5844
):
13
14
.

194.

Manni
A
,
Santen
R
,
Harvey
H
,
Lipton
A
,
Max
D
.
Treatment of breast cancer with gonadotropin-releasing hormone
.
Endocr Rev
.
1986
;
7
(
1
):
89
94
.

195.

Jordan
VC
,
Koerner
S
.
Tamoxifen (ICI 46,474) and the human carcinoma 8S oestrogen receptor
.
Eur J Cancer
.
1975
;
11
(
3
):
205
206
.

196.

Cash
R
,
Brough
AJ
,
Cohen
MN
,
Satoh
PS
.
Aminoglutethimide (Elipten-Ciba) as an inhibitor of adrenal steroidogenesis: mechanism of action and therapeutic trial
.
J Clin Endocrinol Metab
.
1967
;
27
(
9
):
1239
1248
.

197.

Horsley
JS
III,
Newsome
HH
,
Brown
PW
,
Neifeld
JP
,
Terz
JJ
,
Lawrence
W
Jr
.
Medical adrenalectomy in patients with advanced breast cancer
.
Cancer
.
1982
;
49
(
6
):
1145
1149
.

198.

Newsome
HH
,
Brown
PW
,
Terz
JJ
,
Lawrence
W
Jr
.
Medical and surgical adrenalectomy in patients with advanced breast carcinoma
.
Cancer
.
1977
;
39
(
2
):
542
546
.

199.

Newsome
HH
Jr,
Brown
PW
,
Terz
JJ
,
Lawrence
W
Jr
.
Medical adrenalectomy and plasma steroids in advanced breast carcinoma
.
Surgery
.
1978
;
83
(
1
):
83
89
.

200.

Santen
RJ
,
Santner
S
,
Davis
B
,
Veldhuis
J
,
Samojlik
E
,
Ruby
E
.
Aminoglutethimide inhibits extraglandular estrogen production in postmenopausal women with breast carcinoma
.
J Clin Endocrinol Metab
.
1978
;
47
(
6
):
1257
1265
.

201.

Gale
KE
.
Treatment of advanced breast cancer with aminoglutethimide: a 14-year experience. Cancer Res. 1982;42(8 Suppl):3389s–3396s
.

202.

Brodie
AM
,
Schwarzel
WC
,
Shaikh
AA
,
Brodie
HJ
.
The effect of an aromatase inhibitor, 4-hydroxy-4-androstene-3,17-dione, on estrogen-dependent processes in reproduction and breast cancer
.
Endocrinology
.
1977
;
100
(
6
):
1684
1695
.

203.

Brodie
AM
,
Coombes
RC
,
Dowsett
M
.
Aromatase inhibitors: basic and clinical studies
.
J Steroid Biochem
.
1987
;
27
(
4-6
):
899
903
.

204.

Powles
TJ
,
Davey
JB
,
McKinna
A
.
A feasibility trial of tamoxifen chemoprevention of breast cancer in Great Britain
.
Cancer Invest
.
1988
;
6
(
5
):
621
624
.

205.

Key
T
,
Appleby
P
,
Barnes
I
,
Reeves
G
;
Endogenous Hormones and Breast Cancer Collaborative Group
.
Endogenous sex hormones and breast cancer in postmenopausal women: reanalysis of nine prospective studies
.
J Natl Cancer Inst
.
2002
;
94
(
8
):
606
616
.

206.

Kaaks
R
,
Rinaldi
S
,
Key
TJ
,
Berrino
F
,
Peeters
PH
,
Biessy
C
,
Dossus
L
,
Lukanova
A
,
Bingham
S
,
Khaw
KT
,
Allen
NE
,
Bueno-de-Mesquita
HB
,
van Gils
CH
,
Grobbee
D
,
Boeing
H
,
Lahmann
PH
,
Nagel
G
,
Chang-Claude
J
,
Clavel-Chapelon
F
,
Fournier
A
,
Thiébaut
A
,
González
CA
,
Quirós
JR
,
Tormo
MJ
,
Ardanaz
E
,
Amiano
P
,
Krogh
V
,
Palli
D
,
Panico
S
,
Tumino
R
,
Vineis
P
,
Trichopoulou
A
,
Kalapothaki
V
,
Trichopoulos
D
,
Ferrari
P
,
Norat
T
,
Saracci
R
,
Riboli
E
.
Postmenopausal serum androgens, oestrogens and breast cancer risk: the European prospective investigation into cancer and nutrition
.
Endocr Relat Cancer
.
2005
;
12
(
4
):
1071
1082
.

207.

Knight
WA
,
Livingston
RB
,
Gregory
EJ
,
McGuire
WL
.
Estrogen receptor as an independent prognostic factor for early recurrence in breast cancer
.
Cancer Res
.
1977
;
37
(
12
):
4669
4671
.

208.

McGuire
WL
,
Horwitz
KB
,
Zava
DT
,
Garola
RE
,
Chamness
GC
.
Hormones in breast cancer: update 1978
.
Metabolism
.
1978
;
27
(
4
):
487
501
.

209.

McGuire
WL
,
Horwitz
KB
,
De La Garza
M
.
Selecting endocrine therapy in breast cancer
.
Curr Top Mol Endocrinol
.
1976
;
4
:
28
35
.

210.

Fuqua
AW
,
Wiltschke
C
,
Zhang
QX
,
Borg
A
,
Castles
CG
,
Friedrichs
WE
,
Hopp
T
,
Hilsenbeck
S
,
Mohshin
S
,
O’Connell
PO
,
Allred
DC
.
A hypersensitive estrogen receptor-α mutation in premalignant breast lesions
.
Cancer Res
.
2000
;
60
(
15
):
4026
4029
.

211.

Fuqua
SA
.
The role of estrogen receptors in breast cancer metastasis
.
J Mammary Gland Biol Neoplasia
.
2001
;
6
(
4
):
407
417
.

212.

Fuqua
SA
,
Guowei
GU
,
Rechoum
Y
.
Estrogen receptor (ER) α mutations in breast cancer: hidden in plain sight
.
Breast Cancer Res Treat
.
2014
;
144
(
1
):
11
19
.

213.

Patel
HK
,
Bihani
T
.
Selective estrogen receptor modulators (SERMs) and selective estrogen receptor degraders (SERDs) in cancer treatment
.
Pharmacol Ther
.
2018
;
186
:
1
24
.

214.

Boér
K
.
Fulvestrant in advanced breast cancer: evidence to date and place in therapy
[
published correction appears in Ther Adv Med Oncol. 2017;9(11):725
].
Ther Adv Med Oncol
.
2017
;
9
(
7
):
465
479
.

215.

Coen
P
,
Kulin
H
,
Ballantine
T
,
Zaino
R
,
Frauenhoffer
E
,
Boal
D
,
Inkster
S
,
Brodie
A
,
Santen
R
.
An aromatase-producing sex-cord tumor resulting in prepubertal gynecomastia
.
N Engl J Med
.
1991
;
324
(
5
):
317
322
.

216.

Ham
S
,
Meachem
SJ
,
Choong
CS
,
Charles
AK
,
Baynam
GS
,
Jones
TW
,
Samarajeewa
NU
,
Simpson
ER
,
Brown
KA
.
Overexpression of aromatase associated with loss of heterozygosity of the STK11 gene accounts for prepubertal gynecomastia in boys with Peutz-Jeghers syndrome
.
J Clin Endocrinol Metab
.
2013
;
98
(
12
):
E1979
E1987
.

217.

Agarwal
VR
,
Takayama
K
,
Van Wyk
JJ
,
Sasano
H
,
Simpson
ER
,
Bulun
SE
.
Molecular basis of severe gynecomastia associated with aromatase expression in a fibrolamellar hepatocellular carcinoma
.
J Clin Endocrinol Metab
.
1998
;
83
(
5
):
1797
1800
.

218.

Demura
M
,
Martin
RM
,
Shozu
M
,
Sebastian
S
,
Takayama
K
,
Hsu
WT
,
Schultz
RA
,
Neely
K
,
Bryant
M
,
Mendonca
BB
,
Hanaki
K
,
Kanzaki
S
,
Rhoads
DB
,
Misra
M
,
Bulun
SE
.
Regional rearrangements in chromosome 15q21 cause formation of cryptic promoters for the CYP19 (aromatase) gene
.
Hum Mol Genet
.
2007
;
16
(
21
):
2529
2541
.

219.

Shozu
M
,
Sebastian
S
,
Takayama
K
,
Hsu
WT
,
Schultz
RA
,
Neely
K
,
Bryant
M
,
Bulun
SE
.
Estrogen excess associated with novel gain-of-function mutations affecting the aromatase gene
.
N Engl J Med
.
2003
;
348
(
19
):
1855
1865
.

220.

Nagasaki
K
,
Horikawa
R
,
Fujisawa
K
,
Hata
I
,
Shigematsu
Y
,
Tanaka
T
.
A case of female pseudohermaphroditism caused by aromatase deficiency
.
Clin Pediatr Endocrinol
.
2004
;
13
(
1
):
59
64
.

221.

Lin
L
,
Ercan
O
,
Raza
J
,
Burren
CP
,
Creighton
SM
,
Auchus
RJ
,
Dattani
MT
,
Achermann
JC
.
Variable phenotypes associated with aromatase (CYP19) insufficiency in humans
.
J Clin Endocrinol Metab
.
2007
;
92
(
3
):
982
990
.

222.

Marino
R
,
Perez Garrido
N
,
Costanzo
M
,
Guercio
G
,
Juanes
M
,
Rocco
C
,
Ramirez
P
,
Warman
DM
,
Ciaccio
M
,
Pena
G
,
Feyling
JG
,
Miras
M
,
Rivarola
MA
,
Belgorosky
A
,
Saraco
N
.
Five new cases of 46,XX aromatase deficiency: clinical follow-up from birth to puberty, a novel mutation, and a founder effect
.
J Clin Endocrinol Metab
.
2015
;
100
(
2
):
E301
E307
.

223.

Greenblatt
RB
.
Estrogen therapy for postmenopausal females
.
N Engl J Med
.
1965
;
272
(
6
):
305
308
.

224.

Wilson
RA
.
The obsolete menopause
.
Conn Med
.
1963
;
27
:
735
736
.

225.

Wilson
RA
,
Wilson
TA
.
The fate of the nontreated postmenopausal woman: a plea for the maintenance of adequate estrogen from puberty to the grave
.
J Am Geriatr Soc
.
1963
;
11
(
4
):
347
362
.

226.

Wilson
RA
,
Brevetti
RE
,
Wilson
TA
.
Specific procedures for the elimination of the menopause
.
West J Surg, Obstet Gynecol
.
1963
;
71
:
110
121
.

227.

Wilson
RA
.
Feminine Forever. New York, NY: Evans; 1968
.

228.

Rogers
J
.
The menopause
.
N Engl J Med
.
1956
;
254
(
16
):
750
756
.

229.

Rogers
J
.
The menopause
.
N Engl J Med
.
1956
;
254
(
15
):
697
704
.

230.

Santen
RJ
,
Allred
DC
,
Ardoin
SP
,
Archer DF, Boyd N, Braunstein GD, Burger HG, Colditz GA, Davis SR, Gambacciani M, Gower BA, Henderson VW, Jarjour WN, Karas RH, Kleerekoper M, Lobo RA, Manson JE, Marsden J, Martin KA, Martin L, Pinkerton JV, Rubinow DR, Teede H, Thiboutot DM, Utian WH; Endocrine Society. Postmenopausal hormone therapy: an Endocrine Society scientific statement. J Clin Endocrinol Metab. 2010;95(7 Suppl 1):s1–s66.

231.

Stuenkel
CA
,
Davis
SR
,
Gompel
A
,
Lumsden
MA
,
Murad
MH
,
Pinkerton
JV
,
Santen
RJ
.
Treatment of symptoms of the menopause: an Endocrine Society clinical practice guideline
.
J Clin Endocrinol Metab
.
2015
;
100
(
11
):
3975
4011
.

232.

Santen
RJ
,
Stuenkel
CA
,
Davis
SR
,
Pinkerton
JV
,
Gompel
A
,
Lumsden
MA
.
Managing menopausal symptoms and associated clinical issues in breast cancer survivors
.
J Clin Endocrinol Metab
.
2017
;
102
(
10
):
3647
3661
.

233.

Manson
JE
,
Chlebowski
RT
,
Stefanick
ML
,
Aragaki
AK
,
Rossouw
JE
,
Prentice
RL
,
Anderson
G
,
Howard
BV
,
Thomson
CA
,
LaCroix
AZ
,
Wactawski-Wende
J
,
Jackson
RD
,
Limacher
M
,
Margolis
KL
,
Wassertheil-Smoller
S
,
Beresford
SA
,
Cauley
JA
,
Eaton
CB
,
Gass
M
,
Hsia
J
,
Johnson
KC
,
Kooperberg
C
,
Kuller
LH
,
Lewis
CE
,
Liu
S
,
Martin
LW
,
Ockene
JK
,
O’Sullivan
MJ
,
Powell
LH
,
Simon
MS
,
Van Horn
L
,
Vitolins
MZ
,
Wallace
RB
.
Menopausal hormone therapy and health outcomes during the intervention and extended poststopping phases of the Women’s Health Initiative randomized trials
.
JAMA
.
2013
;
310
(
13
):
1353
1368
.

234.

Anderson
GL
,
Chlebowski
RT
,
Aragaki
AK
,
Kuller
LH
,
Manson
JE
,
Gass
M
,
Bluhm
E
,
Connelly
S
,
Hubbell
FA
,
Lane
D
,
Martin
L
,
Ockene
J
,
Rohan
T
,
Schenken
R
,
Wactawski-Wende
J
.
Conjugated equine oestrogen and breast cancer incidence and mortality in postmenopausal women with hysterectomy: extended follow-up of the Women’s Health Initiative randomised placebo-controlled trial
.
Lancet Oncol
.
2012
;
13
(
5
):
476
486
.

235.

Santen
RJ
,
Song
Y
,
Yue
W
,
Wang
JP
,
Heitjan
DF
.
Effects of menopausal hormonal therapy on occult breast tumors
.
J Steroid Biochem Mol Biol
.
2013
;
137
:
150
156
.

236.

Santen
RJ
,
Yue
W
,
Heitjan
DF
.
Occult breast tumor reservoir: biological properties and clinical significance
.
Horm Cancer
.
2013
;
4
(
4
):
195
207
.

237.

Santen
RJ
,
Yue
W
,
Heitjan
DF
.
Modeling of the growth kinetics of occult breast tumors: role in interpretation of studies of prevention and menopausal hormone therapy
.
Cancer Epidemiol Biomarkers Prev
.
2012
;
21
(
7
):
1038
1048
.

238.

Song
RX
,
Mor
G
,
Naftolin
F
,
McPherson
RA
,
Song
J
,
Zhang
Z
,
Yue
W
,
Wang
J
,
Santen
RJ
.
Effect of long-term estrogen deprivation on apoptotic responses of breast cancer cells to 17β-estradiol
.
J Natl Cancer Inst
.
2001
;
93
(
22
):
1714
1723
.

239.

Yue
W
,
Wang
J
,
Atkins
KA
,
Bottalico
L
,
Mesaros
C
,
Blair
IA
,
Santen
RJ.
Effect of a tissue selective estrogen complex on breast cancer: role of unique properties of conjugated equine estrogen
.
Int J Cancer
.
2018
;
143
(
5
):
1259
1268
.

240.

Bender
CM
,
Merriman
JD
,
Gentry
AL
,
Ahrendt
GM
,
Berga
SL
,
Brufsky
AM
,
Casillo
FE
,
Dailey
MM
,
Erickson
KI
,
Kratofil
FM
,
McAuliffe
PF
,
Rosenzweig
MQ
,
Ryan
CM
,
Sereika
SM
.
Patterns of change in cognitive function with anastrozole therapy
.
Cancer
.
2015
;
121
(
15
):
2627
2636
.

241.

Mendelsohn
ME
,
Karas
RH
.
HRT and the young at heart
.
N Engl J Med
.
2007
;
356
(
25
):
2639
2641
.

242.

Shozu
M
,
Simpson
ER
.
Aromatase expression of human osteoblast-like cells
.
Mol Cell Endocrinol
.
1998
;
139
(
1–2
):
117
129
.

243.

Lønning
PE
,
Haynes
BP
,
Straume
AH
,
Dunbier
A
,
Helle
H
,
Knappskog
S
,
Dowsett
M
.
Exploring breast cancer estrogen disposition: the basis for endocrine manipulation
.
Clin Cancer Res
.
2011
;
17
(
15
):
4948
4958
.

244.

Simpson
ER
,
Brown
KA
.
Minireview: obesity and breast cancer: a tale of inflammation and dysregulated metabolism
.
Mol Endocrinol
.
2013
;
27
(
5
):
715
725
.

245.

Morselli
E
,
Santos
RS
,
Criollo
A
,
Nelson
MD
,
Palmer
BF
,
Clegg
DJ
.
The effects of oestrogens and their receptors on cardiometabolic health
.
Nat Rev Endocrinol
.
2017
;
13
(
6
):
352
364
.

246.

Qiu
J
,
Bosch
MA
,
Meza
C
,
Navarro
UV
,
Nestor
CC
,
Wagner
EJ
,
Rønnekleiv
OK
,
Kelly
MJ
.
Estradiol protects proopiomelanocortin neurons against insulin resistance
.
Endocrinology
.
2018
;
159
(
2
):
647
664
.

247.

Wang
HH
,
Zhou
CL
,
Lv
M
,
Yang
Q
,
Li
JX
,
Hou
M
,
Lin
J
,
Liu
XM
,
Wu
YT
,
Sheng
JZ
,
Huang
HF
.
Prenatal high estradiol exposure induces sex-specific and dietarily reversible insulin resistance through decreased hypothalamic INSR
.
Endocrinology
.
2018
;
159
(
1
):
465
476
.

248.

Bedenbaugh
MN
,
D’Oliveira
M
,
Cardoso
RC
,
Hileman
SM
,
Williams
GL
,
Amstalden
M
.
Pubertal escape from estradiol negative feedback in ewe lambs is not accounted for by decreased ESR1 mRNA or protein in kisspeptin neurons
.
Endocrinology
.
2018
;
159
(
1
):
426
438
.

249.

Phumsatitpong
C
,
Moenter
SM
.
Estradiol-dependent stimulation and suppression of gonadotropin-releasing hormone neuron firing activity by corticotropin-releasing hormone in female mice
.
Endocrinology
.
2018
;
159
(
1
):
414
425
.

250.

Weems
P
,
Smith
J
,
Clarke
IJ
,
Coolen
LM
,
Goodman
RL
,
Lehman
MN
.
Effects of season and estradiol on KNDy neuron peptides, colocalization with D2 dopamine receptors, and dopaminergic inputs in the ewe
.
Endocrinology
.
2017
;
158
(
4
):
831
841
.

251.

Royston
SE
,
Yasui
N
,
Kondilis
AG
,
Lord
SV
,
Katzenellenbogen
JA
,
Mahoney
MM
.
ESR1 and ESR2 differentially regulate daily and circadian activity rhythms in female mice
.
Endocrinology
.
2014
;
155
(
7
):
2613
2623
.

252.

Furuta
M
,
Numakawa
T
,
Chiba
S
,
Ninomiya
M
,
Kajiyama
Y
,
Adachi
N
,
Akema
T
,
Kunugi
H
.
Estrogen, predominantly via estrogen receptor α, attenuates postpartum-induced anxiety- and depression-like behaviors in female rats
.
Endocrinology
.
2013
;
154
(
10
):
3807
3816
.

253.

Miller
NR
,
Jover
T
,
Cohen
HW
,
Zukin
RS
,
Etgen
AM
.
Estrogen can act via estrogen receptor α and β to protect hippocampal neurons against global ischemia-induced cell death
.
Endocrinology
.
2005
;
146
(
7
):
3070
3079
.

254.

Lund
TD
,
Rovis
T
,
Chung
WC
,
Handa
RJ
.
Novel actions of estrogen receptor-β on anxiety-related behaviors
.
Endocrinology
.
2005
;
146
(
2
):
797
807
.

255.

Vanacker
C
,
Moya
MR
,
DeFazio
RA
,
Johnson
ML
,
Moenter
SM
.
Long-term recordings of arcuate nucleus kisspeptin neurons reveal patterned activity that is modulated by gonadal steroids in male mice
.
Endocrinology
.
2017
;
158
(
10
):
3553
3564
.

256.

Minabe
S
,
Ieda
N
,
Watanabe
Y
,
Inoue
N
,
Uenoyama
Y
,
Maeda
KI
,
Tsukamura
H
.
Long-term neonatal estrogen exposure causes irreversible inhibition of LH pulses by suppressing arcuate kisspeptin expression via estrogen receptors α and β in female rodents
.
Endocrinology
.
2017
;
158
(
9
):
2918
2929
.

257.

Hu
P
,
Liu
J
,
Yasrebi
A
,
Gotthardt
JD
,
Bello
NT
,
Pang
ZP
,
Roepke
TA
.
Gq Protein-coupled membrane-initiated estrogen signaling rapidly excites corticotropin-releasing hormone neurons in the hypothalamic paraventricular nucleus in female mice
.
Endocrinology
.
2016
;
157
(
9
):
3604
3620
.

258.

Farman
HH
,
Windahl
SH
,
Westberg
L
,
Isaksson H, Egecioglu E, Schele E, Ryberg H, Jansson JO, Tuukkanen J, Koskela A, Xie SK, Hahner L, Zehr J, Clegg DJ, Lagerquist MK, Ohlsson C. Female mice lacking estrogen receptor-α in hypothalamic proopiomelanocortin (POMC) neurons display enhanced estrogenic response on cortical bone mass. Endocrinology. 2016;157(8):3242–3252.

259.

Grissom
EM
,
Daniel
JM
.
Evidence for ligand-independent activation of hippocampal estrogen receptor-α by IGF-1 in hippocampus of ovariectomized rats
.
Endocrinology
.
2016
;
157
(
8
):
3149
3156
.

260.

Ferasyi
TR
,
Barrett
PH
,
Blache
D
,
Martin
GB
.
Modeling the male reproductive endocrine axis: potential role for a delay mechanism in the inhibitory action of gonadal steroids on GnRH pulse frequency
.
Endocrinology
.
2016
;
157
(
5
):
2080
2092
.

261.

Stephens
SB
,
Chahal
N
,
Munaganuru
N
,
Parra
RA
,
Kauffman
AS
.
Estrogen stimulation of Kiss1 expression in the medial amygdala involves estrogen receptor-α but not estrogen receptor-β
.
Endocrinology
.
2016
;
157
(
10
):
4021
4031
.

262.

Dubois
SL
,
Wolfe
A
,
Radovick
S
,
Boehm
U
,
Levine
JE
.
Estradiol restrains prepubertal gonadotropin secretion in female mice via activation of ERα in kisspeptin neurons
.
Endocrinology
.
2016
;
157
(
4
):
1546
1554
.

263.

Ruka
KA
,
Burger
LL
,
Moenter
SM
.
both estrogen and androgen modify the response to activation of neurokinin-3 and κ-opioid receptors in arcuate kisspeptin neurons from male mice
.
Endocrinology
.
2016
;
157
(
2
):
752
763
.

264.

Shults
CL
,
Pinceti
E
,
Rao
YS
,
Pak
TR
.
Aging and loss of circulating 17β-estradiol alters the alternative splicing of ERβ in the female rat brain
.
Endocrinology
.
2015
;
156
(
11
):
4187
4199
.

265.

Sárvári
M
,
Kalló
I
,
Hrabovszky
E
,
Solymosi
N
,
Rodolosse
A
,
Vastagh
C
,
Auer
H
,
Liposits
Z
.
Hippocampal gene expression is highly responsive to estradiol replacement in middle-aged female rats
.
Endocrinology
.
2015
;
156
(
7
):
2632
2645
.

266.

Kenealy
BP
,
Keen
KL
,
Garcia
JP
,
Richter
DJ
,
Terasawa
E
.
Prolonged infusion of estradiol benzoate into the stalk median eminence stimulates release of GnRH and kisspeptin in ovariectomized female rhesus macaques
.
Endocrinology
.
2015
;
156
(
5
):
1804
1814
.

267.

Mittelman-Smith
MA
,
Wong
AM
,
Kathiresan
AS
,
Micevych
PE
.
Classical and membrane-initiated estrogen signaling in an in vitro model of anterior hypothalamic kisspeptin neurons
.
Endocrinology
.
2015
;
156
(
6
):
2162
2173
.

268.

Dubois
SL
,
Acosta-Martínez
M
,
DeJoseph
MR
,
Wolfe
A
,
Radovick
S
,
Boehm
U
,
Urban
JH
,
Levine
JE
.
Positive, but not negative feedback actions of estradiol in adult female mice require estrogen receptor α in kisspeptin neurons
.
Endocrinology
.
2015
;
156
(
3
):
1111
1120
.

269.

Barron
AM
,
Brown
MA
,
Morgan
TE
,
Pike
CJ
.
Impact of continuous versus discontinuous progesterone on estradiol regulation of neuron viability and sprouting after entorhinal cortex lesion in female rats
.
Endocrinology
.
2015
;
156
(
3
):
1091
1099
.

270.

Milne
MR
,
Haug
CA
,
Ábrahám
IM
,
Kwakowsky
A
.
Estradiol modulation of neurotrophin receptor expression in female mouse basal forebrain cholinergic neurons in vivo
.
Endocrinology
.
2015
;
156
(
2
):
613
626
.

271.

Almey
A
,
Cannell
E
,
Bertram
K
,
Filardo
E
,
Milner
TA
,
Brake
WG
.
Medial prefrontal cortical estradiol rapidly alters memory system bias in female rats: ultrastructural analysis reveals membrane-associated estrogen receptors as potential mediators
.
Endocrinology
.
2014
;
155
(
11
):
4422
4432
.

272.

Yeo
SH
,
Herbison
AE
.
Estrogen-negative feedback and estrous cyclicity are critically dependent upon estrogen receptor-α expression in the arcuate nucleus of adult female mice
.
Endocrinology
.
2014
;
155
(
8
):
2986
2995
.

273.

Chen
C
,
Kuo
J
,
Wong
A
,
Micevych
P
.
Estradiol modulates translocator protein (TSPO) and steroid acute regulatory protein (StAR) via protein kinase A (PKA) signaling in hypothalamic astrocytes
.
Endocrinology
.
2014
;
155
(
8
):
2976
2985
.

274.

Arimoto
JM
,
Wong
A
,
Rozovsky
I
,
Lin
SW
,
Morgan
TE
,
Finch
CE
.
Age increase of estrogen receptor-α (ERα) in cortical astrocytes impairs neurotrophic support in male and female rats
.
Endocrinology
.
2013
;
154
(
6
):
2101
2113
.

275.

MacLusky
NJ
.
Understanding the direct synaptic effects of estradiol
.
Endocrinology
.
2013
;
154
(
2
):
581
583
.

276.

Witty
CF
,
Gardella
LP
,
Perez
MC
,
Daniel
JM
.
Short-term estradiol administration in aging ovariectomized rats provides lasting benefits for memory and the hippocampus: a role for insulin-like growth factor-I
.
Endocrinology
.
2013
;
154
(
2
):
842
852
.

277.

Clarke
SD
,
Clarke
IJ
,
Rao
A
,
Evans
RG
,
Henry
BA
.
Differential effects of acute and chronic estrogen treatment on thermogenic and metabolic pathways in ovariectomized sheep
.
Endocrinology
.
2013
;
154
(
1
):
184
192
.

278.

Santollo
J
,
Marshall
A
,
Daniels
D
.
Activation of membrane-associated estrogen receptors decreases food and water intake in ovariectomized rats
.
Endocrinology
.
2013
;
154
(
1
):
320
329
.

279.

Spampinato
SF
,
Merlo
S
,
Molinaro
G
,
Battaglia
G
,
Bruno
V
,
Nicoletti
F
,
Sortino
MA
.
Dual effect of 17β-estradiol on NMDA-induced neuronal death: involvement of metabotropic glutamate receptor 1
.
Endocrinology
.
2012
;
153
(
12
):
5940
5948
.

280.

Yang
JA
,
Stires
H
,
Belden
WJ
,
Roepke
TA
.
The arcuate estrogen-regulated transcriptome: estrogen response element-dependent and -independent signaling of ERα in female mice
.
Endocrinology
.
2017
;
158
(
3
):
612
626
.

281.

Vinel
A
,
Hay
E
,
Valera
MC
,
Buscato
M
,
Adlanmerini
M
,
Guillaume
M
,
Cohen-Solal
M
,
Ohlsson
C
,
Lenfant
F
,
Arnal
JF
,
Fontaine
C
.
Role of ERα in the effect of estradiol on cancellous and cortical femoral bone in growing female mice
.
Endocrinology
.
2016
;
157
(
6
):
2533
2544
.

282.

Zhang
Z
,
Liu
J
,
Veldhuis-Vlug
AG
,
Su
Y
,
Foppen
E
,
van der Eerden
BC
,
Koedam
M
,
Bravenboer
N
,
Kalsbeek
A
,
Boelen
A
,
Fliers
E
,
Bisschop
PH
.
Effects of chronic estrogen administration in the ventromedial nucleus of the hypothalamus (VMH) on fat and bone metabolism in ovariectomized rats
.
Endocrinology
.
2016
;
157
(
12
):
4930
4942
.

283.

Connelly
KJ
,
Larson
EA
,
Marks
DL
,
Klein
RF
.
Neonatal estrogen exposure results in biphasic age-dependent effects on the skeletal development of male mice
.
Endocrinology
.
2015
;
156
(
1
):
193
202
.

284.

Pepe
GJ
,
Maniu
A
,
Aberdeen
G
,
Lynch
TJ
,
Albrecht
ED
.
Estrogen regulation of fetal adrenal cortical zone-specific development in the nonhuman primate impacts adrenal production of androgen and cortisol and response to ACTH in females in adulthood
.
Endocrinology
.
2016
;
157
(
5
):
1905
1913
.

285.

Babischkin
JS
,
Aberdeen
GW
,
Pepe
GJ
,
Albrecht
ED
.
Estrogen suppresses interaction of melanocortin 2 receptor and its accessory protein in the primate fetal adrenal cortex
.
Endocrinology
.
2016
;
157
(
12
):
4588
4601
.

286.

Dumitrescu
A
,
Aberdeen
GW
,
Pepe
GJ
,
Albrecht
ED
.
Placental estrogen suppresses cyclin D1 expression in the nonhuman primate fetal adrenal cortex
.
Endocrinology
.
2014
;
155
(
12
):
4774
4784
.

287.

Caroccia
B
,
Seccia
TM
,
Campos
AG
,
Gioco
F
,
Kuppusamy
M
,
Ceolotto
G
,
Guerzoni
E
,
Simonato
F
,
Mareso
S
,
Lenzini
L
,
Fassina
A
,
Rossi
GP
.
GPER-1 and estrogen receptor-β ligands modulate aldosterone synthesis
.
Endocrinology
.
2014
;
155
(
11
):
4296
4304
.

288.

Qi
X
,
Salem
M
,
Zhou
W
,
Sato-Shimizu
M
,
Ye
G
,
Smitz
J
,
Peng
C
.
Neurokinin B exerts direct effects on the ovary to stimulate estradiol production
.
Endocrinology
.
2016
;
157
(
9
):
3355
3365
.

289.

Toda
K
,
Hayashi
Y
,
Ono
M
,
Saibara
T
.
Characterization of ovarian responses to equine chorionic gonadotropin of aromatase-deficient mice with or without 17β-estradiol supplementation
.
Endocrinology
.
2016
;
157
(
5
):
2093
2103
.

290.

Van Kempen
TA
,
Gorecka
J
,
Gonzalez
AD
,
Soeda
F
,
Milner
TA
,
Waters
EM
.
Characterization of neural estrogen signaling and neurotrophic changes in the accelerated ovarian failure mouse model of menopause
.
Endocrinology
.
2014
;
155
(
9
):
3610
3623
.

291.

Moore
AM
,
Prescott
M
,
Campbell
RE
.
Estradiol negative and positive feedback in a prenatal androgen-induced mouse model of polycystic ovarian syndrome
.
Endocrinology
.
2013
;
154
(
2
):
796
806
.

292.

Payrits
M
,
Sághy
É
,
Cseko
K
,
Pohóczky
K
,
Bölcskei
K
,
Ernszt
D
,
Barabás
K
,
Szolcsányi
J
,
Ábrahám
IM
,
Helyes
Z
,
Szoke
É
.
Estradiol sensitizes the transient receptor potential vanilloid 1 receptor in pain responses
.
Endocrinology
.
2017
;
158
(
10
):
3249
3258
.

293.

Yamagata
K
,
Sugimura
M
,
Yoshida
M
,
Sekine
S
,
Kawano
A
,
Oyamaguchi
A
,
Maegawa
H
,
Niwa
H
.
Estrogens exacerbate nociceptive pain via up-regulation of TRPV1 and ANO1 in trigeminal primary neurons of female rats
.
Endocrinology
.
2016
;
157
(
11
):
4309
4317
.

294.

Ghorbanpoor
S
,
Garcia-Segura
LM
,
Haeri-Rohani
A
,
Khodagholi
F
,
Jorjani
M
.
Aromatase inhibition exacerbates pain and reactive gliosis in the dorsal horn of the spinal cord of female rats caused by spinothalamic tract injury
.
Endocrinology
.
2014
;
155
(
11
):
4341
4355
.

295.

Greaves
E
,
Collins
F
,
Esnal-Zufiaurre
A
,
Giakoumelou
S
,
Horne
AW
,
Saunders
PT
.
Estrogen receptor (ER) agonists differentially regulate neuroangiogenesis in peritoneal endometriosis via the repellent factor SLIT3
.
Endocrinology
.
2014
;
155
(
10
):
4015
4026
.

296.

Zhao
Y
,
Park
S
,
Bagchi
MK
,
Taylor
RN
,
Katzenellenbogen
BS
.
The coregulator, repressor of estrogen receptor activity (REA), is a crucial regulator of the timing and magnitude of uterine decidualization
.
Endocrinology
.
2013
;
154
(
3
):
1349
1360
.

297.

Whirledge
S
,
Cidlowski
JA
.
Estradiol antagonism of glucocorticoid-induced GILZ expression in human uterine epithelial cells and murine uterus
.
Endocrinology
.
2013
;
154
(
1
):
499
510
.

298.

Inada
A
,
Fujii
NL
,
Inada
O
,
Higaki
Y
,
Furuichi
Y
,
Nabeshima
YI
.
Effects of 17β-Estradiol and androgen on glucose metabolism in skeletal muscle
.
Endocrinology
.
2016
;
157
(
12
):
4691
4705
.

299.

Litwak
SA
,
Wilson
JL
,
Chen
W
,
Garcia-Rudaz
C
,
Khaksari
M
,
Cowley
MA
,
Enriori
PJ
.
Estradiol prevents fat accumulation and overcomes leptin resistance in female high-fat diet mice
.
Endocrinology
.
2014
;
155
(
11
):
4447
4460
.

300.

Garrido
P
,
Morán
J
,
Alonso
A
,
González
S
,
González
C
.
17β-Estradiol activates glucose uptake via GLUT4 translocation and PI3K/Akt signaling pathway in MCF-7 cells
.
Endocrinology
.
2013
;
154
(
6
):
1979
1989
.

301.

Jelenik
T
,
Roden
M
.
How estrogens prevent from lipid-induced insulin resistance
.
Endocrinology
.
2013
;
154
(
3
):
989
992
.

302.

Camporez
JP
,
Jornayvaz
FR
,
Lee
HY
,
Kanda
S
,
Guigni
BA
,
Kahn
M
,
Samuel
VT
,
Carvalho
CR
,
Petersen
KF
,
Jurczak
MJ
,
Shulman
GI
.
Cellular mechanism by which estradiol protects female ovariectomized mice from high-fat diet-induced hepatic and muscle insulin resistance
.
Endocrinology
.
2013
;
154
(
3
):
1021
1028
.

303.

Nanjappa
MK
,
Hess
RA
,
Medrano
TI
,
Locker
SH
,
Levin
ER
,
Cooke
PS
.
Membrane-localized estrogen receptor 1 is required for normal male reproductive development and function in mice
.
Endocrinology
.
2016
;
157
(
7
):
2909
2919
.

Supplementary data