Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Jan 31.
Published in final edited form as: Nat Aging. 2024 Dec 13;4(12):1731–1744. doi: 10.1038/s43587-024-00767-0

Exploring the effects of estrogen deficiency and aging on organismal homeostasis during menopause

Celine Camon 1,2, Michael Garratt 1, Stephanie M Correa 2
PMCID: PMC11785355  NIHMSID: NIHMS2041190  PMID: 39672893

Abstract

Sex hormone signalling declines during aging, from early mid-life through menopause, as a consequence of reduced circulating estrogens and decreased receptiveness to these hormones in target tissues. Estrogens preserve energy homeostasis and promote metabolic health via coordinated and simultaneous effects throughout the brain and body. Age-associated loss of estrogen production during menopause has been implicated in a higher risk for metabolic diseases and increased mortality. However, it remains unclear whether age-associated changes in homeostasis are dependent on reduced estrogen signalling during menopause. While menopausal hormone therapies containing estrogens can alleviate symptoms, concerns about the risks involved have contributed to a broad decline in the use of these approaches. Non-hormonal therapies have emerged that target tissues or pathways with varying levels of selectivity reduced risk. We summarize here the broad effects of estrogen loss on homeostasis during menopause, current and emerging therapies, and opportunities for understanding homeostatic disruptions associated with menopause.

Introduction

Aging is a principal risk factor for a number of diseases, including cancers, cardiovascular and neurodegenerative conditions1. Circulating sex hormones produced by the ovaries decline during aging (see Box 1 for an overview of estrogen production and signalling)2. Energy homeostasis can be broadly defined as the interplay between energy intake and energy expenditure. When imbalanced, changes in body weight and adiposity often occur, which can lead to metabolic dysfunction and increased risk of mortality and a variety of diseases. Energy homeostasis is maintained not only by peripheral cues from the body, but also by the integration of the central nervous system, specifically the hypothalamus3. Estrogens play a potent role in energy homeostasis across the lifespan and their decline during aging has been implicated in the increased risk for neurological and metabolic diseases 4. The use of estrogens as a treatment to mitigate these effects has been implemented over the last century as menopausal hormone therapies (previously referred to as hormone replacement therapies) and sex hormone replacement. In this review we present an overview of the central and peripheral roles of estrogens in governing homeostasis across the lifespan. Unless otherwise stated, our reference to estrogens refers to the hormone family as a whole rather than one specific member. A number of studies cited in this review do not include how they determine sex or gender. We therefore interpret their definition of women to include those assigned female at birth, individuals who self-report as women or clinical studies focusing on cis women. We recognize that this excludes other individuals who go through menopause and women who do not go through menopause.

Box 1: An overview of Estrogen Actions.

Estrogens are members of the steroid family of hormones derived from cholesterol following a series of metabolic conversions 5. The three main forms of endogenous estrogens are estrone (E1) estradiol (E2) and estriol (E3). The predominant endogenous circulating estrogen in cycling females is 17-beta estradiol (17βE2), produced by the ovaries 6. Estradiol is also synthesized in the testes and is produced by the aromatization of androgens in other tissues 7. Estrogenic effects in the control of both female and male reproduction have been well documented, but estrogens also play key roles in cardiovascular, neuroendocrine, skeletal and metabolism physiology 8.

The molecular actions of most estrogens are mediated by estrogen receptors (ERs). The classical ERs include estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ), members of the nuclear receptor family. While both receptors share many conserved domains, ligand binding to either receptor can have distinct physiological roles, depending on the tissue or site of interaction 9. ERs are expressed in various tissues throughout the body, including in reproductive (uterus, ovaries, breast, vagina, prostate and epididymis) and non-reproductive (lungs, heart, liver, colon and brain) organs 10. Within the brain of multiple species, ERα is expressed in the amygdala, hippocampus, cortex, midbrain, brainstem, pituitary gland, preoptic area and the hypothalamus 1113. ERβ is also expressed throughout the brain, with abundant levels in the hippocampus and hypothalamus 14.

The classical mechanism of estrogen signalling involves the following:

  • Diffusion of estrogens across the plasma membrane

  • Subsequent binding to the N terminus of the estrogen receptor in the cytoplasm

  • Confirmational change of the ligand-receptor complex

  • Translocation of the ligand-receptor complex to the nucleus

  • Transcription of targeted genes by binding to DNA sequences known as estrogen response elements (EREs) 15,16.

Estrogens can also signal by binding to membrane bound G-protein coupled receptors (GPERs), increasing intracellular calcium and activating signalling cascades such as MAPK 17,18.

Estrogen deficiency disrupts organismal homeostasis

A notable change that occurs in estrogen signalling over human aging is the menopause transition, which is driven by changes in the follicular reserve of the ovaries. Ovarian aging is principally classified by a decline in both the quantity and quality of oocytes, with primordial follicles gradually reducing from millions at birth to within hundreds by perimenopause 19,20. The depleted ovarian reserve and reduced production of estrogens and progestogens from the ovaries, results in wide-ranging changes in menstrual length and altered cycling of HPG axis hormones during perimenopause, followed by a cessation of menstruation and a gradual reduction of circulating estrogens to negligible levels post-menopause 21,22. The capability to reproduce is lost after menopause at the average age of 51 23. Over this period changes in body composition occur including an increase in adiposity, although this increase in body fat is generally in line with the gradual increase in adiposity that occurs progressively across life 24,25. The risk of cardiovascular events increases after menopause, as estrogens are thought to act protectively against cardiovascular disease by influencing arterial vasodilation and vascular tone 26,27. By contrast, bone density shows a marked decline after menopause compared to earlier ages, with this reduction beginning many years before the onset of menopause 28,29 and the risk for fractures is increased post-menopause30. An increase in bone turnover with reduced bone formation results in the loss of trabecular bone and overall bone density 31. Along with a loss of estrogens, a variety of hormones show dramatic changes in their circulating levels over and after menopause. For example, increased FSH is implicated in driving the loss of bone mass that co-occurs with lowered circulating estrogen 32. However, estrogen-based menopausal hormone therapy (MHT) can ameliorate some of the changes in body composition that occur with menopause (discussed below), suggesting that estrogen signalling, both directly or via effects on other aspects of the HPG-axis (e.g. reducing FSH levels), is implicated in the changes in organismal homeostasis that occur over menopause (Figure 1).

Figure 1:

Figure 1:

Overview of estrogen signalling and physiological effects. 17-Beta Estradiol (17βE2) is the predominant circulating estrogen produced by the ovary following the release of FSH and LH from the anterior pituitary. a) 17βE2 binds to classical estrogen receptors, translocates to the nucelus and binds to estrogen response elements (EREs) which increases gene transcription influencing metabolism, anti-inflammatory pathways and mitochondrial activation by the nuclear initiated pathway. b) 17βE2 binding to membrane bound G protein coupled receptors initiate calcium signalling, cAMP, PKA and eNOS pathways. c) Estrogen signalling plays a role in energy balance, thermoregulation, bone and muscle growth and is essential for reproductive behaviours. d) reduced circulating estrogens post menopause increases the risk for cardiovascular disease (CVD), metabolic dysfunction, thermodysregulatinon, osteoporosis, neurological disorders, bone degeneration and sleep loss.

Evidence of ovarian estrogen loss as the driver of changes in health during menopause is observable in comparisons of those who have undergone surgical removal of their ovaries for health conditions. Premenopausal research participants undergoing oophorectomy for benign health conditions have been compared to reference groups who have not undergone a surgical sterilization procedure, or who have undergone a hysterectomy with ovarian conservation 33,34. Ovary removal has been associated with an increased cardiovascular disease incidence/severity 35, increased mortality 33,35, increased risk of bone fracture 36, increased risk of dementia37, and increased anxiety, 38 although it has been noted that population differences and the reasons for why people undergo a specific surgery could explain these relationships. Indeed, in large-scale comparisons (~25,000 individuals) of women who entered the Women’s Health Initiative observational study, no long-term negative health consequences of ovarian removal were documented, although most went through surgery around the age of typical menopause 39. Overall, studies have indicated that risk of impaired health with oophorectomy is greatest when ovaries are removed early in life, before the menopause transition begins. For example, worsening of vasomotor symptoms and a decline in sexual function and satisfaction have been reported in patients who underwent an oophrectomy while in perimenopause 40. This is consistent with health consequences of natural menopause, which appear to be more profound when menopause occurs prematurely 41. Thus, abrupt and/or early loss of ovarian estrogen production appears to be associated with the most profound health effects in humans, although changes in associated hormones or confounding disease states may explain these results.

Estrogen replacement can extend lifespan in animal models

The direct effects of ovarian hormone loss on energy homeostasis have been studied in rodent models. Ovariectomy (OVX) in mice and rats is considered an obesogenic manipulation since this is associated with increased body weight and adiposity after surgery 42,43. This is counteracted by treatment with 17βE2, highlighting the importance of estrogens in regulation of body weight in rodents 44,45. Similar effects have been noted in clinical studies, with participants randomised to MHT containing estrogens over four years demonstrating a lower BMI in comparison with placebo participants 46. Effects of OVX on lifespan and aging have been less consistent. If endogenous estrogen production has a positive effect on female survival then survival should be consistently reduced with OVX. Some studies show that OVX can reduce survival 46,47, others that OVX increases lifespan 47,48 with others demonstrating little change in survival 49,50. Notably, many of these experiments were conducted prior to 1980 when deaths due to infectous disease where often reported in laboratory housed rodents and the average lifespan was much shorter, thus some results maybe complicated by effects of estrogens on the immune system.

There has been a more recent focus on the effects of direct estrogen treatment on lifespan in animal models. The Interventions Testing Program, funded by the National Institute of Aging, has reported that treatment with 17-alpha estradiol (17αE2), a steroisomer of 17βE2, can increase the lifespan of male mice by nearly 20%, although the same treatment has no effect on the survival of females 51,52. Male mice treated with 17αE2 have improved glucose tolerance 5355 and greater physical function and muscle mass at later ages 56, suggesting that this treatment slows various aspects of aging. The pathway through which 17αE2 is acting to influence aging is unclear, although 17αE2 treatment stimulates ERα expression in vitro, and the metabolic benefits of this treatment for mice on a high fat diet are lost in mice that do not express ERα in all tissues 57. It has been assumed that at least a part of the metabolic and anti-aging benefits of this treatment are centrally mediated by ERα signalling in the hypothalamus, which regulates changes in feeding behaviour and energy expenditure (explained below). Indeed, mice that have a knockdown of pro-opiomelanocortin (Pomc), a precursor polypeptide that controls feeding behaviour, show a diminished response to 17αE2 for some traits 58. However, a recent study has demonstrated that changes in body weight and glucose tolerance with 17αE2 treatment when on a high fat diet ae maintained even when ERα expression is strongly reduced in the hypothalamus through loss of expression in either glutamatergic or GABAergic neurons 55. It is possible that ERα signalling in multiple neuronal populations is required to drive metabolic changes in response to 17αE2, or that peripheral estrogen receptors are important in causing metabolic health benefits.

There has also been an interest in understanding why 17αE2 provides anti-aging benefits only to male and not female mice. Initial assumptions were that 17αE2 is replicating in male mice an anti-aging benefit that female mice receive from circulating 17βE2. Indeed, it is hypothesized that individuals sexed female on average live longer than individuals sexed male as a consequence of their exposure to ovarian estrogens and therefore understanding how 17αE2 acts to improve male survival could also help to explain sexual dimorphism in aging 59,60. Suprisingly, however, the benefical effects of 17αE2 on glucose tolerance and physical function depend on male mice having intact testes, with castrated animals not showing the anti-aging benefits of 17αE2 treatment 53,56. OVX female mice also show limited responsiveness to 17αE2 53,56, suggesting that this treatment is not simply replacing the benefical effects that female animals gain from ovarian estrogen production. This work highlights the importance of estrogen signalling in male aging and a role for ERα in providing the health benefits of treatment, although the precise signalling pathways remain unclear. A similar 15% increase in lifespan has recently been reported in male mice treated with estriol, demonstrating that the lifespan response is not specific to 17αE2 and multiple estrogens can elicit the anti-aging response in male mice. Interestingly however, the same treatment caused a 6% reduction in female lifespan in mice that had inctact ovaries for the course of treatment 61.

While 17αE2 fails to influence aging in female mice, the impact of ovaries on female aging has been illustrated via ovarian transplant studies in CBA mice, which show ovarian failure relatively early in life compared to other mouse strains. In these studies OVX alone had a negligible effect on survival compared to controls, but surgical transplantation of young ovaries to middle-aged mice (11 months old) increased life-expectancy 62,63. Female mice with transplanted ovaries also showed less evidence of cardiomyopathy at death compared to controls, and this benefit was greatest in animals that were still experiencing reproductive cycles at the time they received a young ovary 64. This is consistent with the hypothesis that MHT may provide the greatest health benefits when initiated at the time of the menopause transition (discussed in further detail later). However, while it was initially assumed that the benefits of receiving young ovaries at mid-life was a consequence of increased exposure to ovarian hormones, particularly estradiol, more recent studies suggest that these benefits are hormone-independent and are a consequence of the transfer of somatic cells in ovaries. Selectively destroying germ cells from ovaries, which precludes follicle development and estrogen production, before transplanting them to recipients failed to block the lifespan effect and instead surprisingly increased the degree of lifespan extension 65. Notably, this was a small study (n=6 per group) that only assessed changes in median lifespan in a model with early ovarian senescence and relatively short longevity. However, the results would suggest that other aspects of ovarian tissue, in addition to germline cells and associated estrogen production, may be important in the control of female lifespan.

Prior research in humans and animal models highlights a key role of ovarian hormones, and likely ovarian estradiol production, in the control of mortality and changes in energy homeostasis across the life course. However, to date there has been virtually no research into how estrogen signalling through specific receptors or tissues influences lifespan directly, and most results are complicated by the impact that broad manipulation of the ovary/circulating estrogens has on associated hormones (e.g. FSH, LH, activins). Instead, there has been a focus over the last decade on how estrogen signalling influences organismal energy homeostasis, which has increased the understanding of how estrogen signalling through specific receptors in specific cell types influences different aspects of activity, food intake and energy expenditure.

Estrogen deficiency results in coordinated metabolic disruptions across the brain and body

Strong evidence that estrogen signalling can benefit metabolic health comes from rodent studies. Similar to changes in caloric intake and physical activity across the menstrual cycle in humans 66,67, cyclic patterns of feeding, physical activity, and body temperature have been reported across the estrous cycle in rats 68,69 and mice 7072. Although these effects could be mediated by multiple hormones from the ovaries or from other signals produced in response to ovarian secretions, data from genetically engineered knockout mice point to a major role of estrogen signalling through estrogen receptor alpha (ERα). Eliminating ERα either globally or in the central nervous system leads to obesity due to increased feeding, reduced movement, and reduced thermogenesis in both males and females 7375, pointing to a critical role for ERα signalling in energy balance.

Tissue-specific or cell-type-specific conditional knockout mouse models have begun to reveal the central mediators of estrogen’s metabolic effects. Estrogens modulate food intake via ERα signalling in the arcuate nucleus 71,75 and the nucleus of the solitary tract 76, as well as through other estrogen receptors in the arcuate 77. Central ERα signalling also has potent effects on energy expenditure. Estrogens modulate two types of energy expenditure via ERα signalling in the ventromedial nucleus of the hypothalamus 75,7881 and the medial preoptic area 82,83. Interestingly, the metabolic effects of estrogens via the hypothalamus (and the preoptic area, which is functionally related to the hypothalamus but has a distinct developmental origin 84) are female-specific. ERα signalling outside of the hypothalamus, in the medial amygdala, alters physical activity in male mice 85. Together these studies suggest that estrogens in the non-masculinized brain act on multiple regions in concert to maintain energy balance, and on distinct regions in the masculinized brain to exert a similar effect.

Aging is very likely to alter both estrogen signalling and cellular processes in the hypothalamus. The expression of ERα and ERβ decline with age in the brains of female rats, although this occurs in a region-specific manner 86,87. These age-effects are also observable in OVX rats treated with estradiol which may suggest that old and young animals will show different signalling responses to estrogen treatment because they differ in receptor expression 86. In support of this, previous research in rats has shown that old animals require greater estradiol doses to elicit memory improvements when compared to younger animals 88. More generally, many cellular processes within hypothalamic cells are altered with aging. Gene expression analyses comparing aged and young female mice identified differences in aging signatures within cell clusters with predicted roles in feeding and energy balance 89. These signatures of hypothalamic aging may interact with the effects of estrogens on metabolism. Indeed, knocking out the G-protein coupled estrogen receptor (GPER) can interfere with metabolic health later in life. Female mice lacking GPER globally have higher adiposity 90 but the phenotype has been reported to emerge after 16 weeks of age 91.

Estrogen actions in peripheral tissues are also potent mediators of energy homeostasis, largely shown in rodents but also in human studies. Both ERs are expressed in the mouse liver, with ERα being the most abundant 92,93. ERα in the liver plays an important role in glucose and insulin like growth factor (IGF-1) signalling in rodents 92,94. Polymorphisms of ESR1, the gene that encodes ERα, are associated with increased plasma glucose levels and type II diabetes in human patients, demonstrating the important role of estrogen signalling in metabolic homeostasis 95. ERα agonists can ameliorate steatosis in the liver of aromatase knockout male mice, while ablation of ERα in the liver leads to increased lipid and triglyceride accumulation in both male and female mice 94,96,97. The potential of estrogen to enhance liver regeneration has been discussed for decades, with both estradiol treatment and ERα binding activity supporting the induction of regrowth post hepatectomy in rodents 98,99. Interestingly, oral estrogen administration after menopause in humans with non-alcoholic fatty liver disease actually worsened the disease, however, transdermal routes reduced the prevalence 100. It is likely that while estrogen signalling plays an important role in maintaining homeostasis in the liver, the delivery route and timing of treatment may influence its protective potential.

Adipose tissue is largely considered an endocrine gland with insulin sensitivity, immune responses and mitochondrial activity being labelled as the hallmarks of adipocyte physiology 4. Adiposity deposition changes after menopause, redistributing from femoral and gluteal subcutaneous deposits to increased visceral adipose tissue. This elevated central adiposity can increase insulin resistance, lipolysis and lead to overall metabolic disease 4,101. It is widely known that OVX in rodents induces weight gain, with mice showing an increase in abdominal fat after 12 weeks 102. In OVX mice, fat deposition changes as a direct consequence of reduced estrogen signalling, with increased visceral and subcutaneous fat that is not due to a reduction in physical activity 103. OVX also increases adiponectin levels in female rats, a hormone secreted by adipose tissue 104. Global ERαKO mice and selective knockdown of ERα in adipose tissue and adipocytes significantly increases body fat, adipocyte size, tissue inflammation and fibrosis in both male and female animals 105.

White (WAT) and brown (BAT) adipose tissues have distinct metabolic functions, therefore it is important to discuss their roles separately when considering fluctuating estrogen levels. WAT is essential for glucose homeostasis, the secretion of leptin and adiponectin and energy storage, while BAT plays a major role in thermogenesis and energy expenditure 106109. Post-menopausal persons have a decreased core temperature and interestingly, a reduction in core temperature has been linked with large decreases in energy expenditure 110,111. The conversion of WAT to BAT known as browning, which can happen in certain WAT depots, is generally considered protective against metabolic dysfunction, due to BAT’s capacity to consume glucose and fatty acids 112. The role of estrogen signalling in beiging is somewhat controversial. The addition of 17βE2 to OVX rats leads to increased expression of thermogenesis markers such as the UCP-1 protein and increased temperature at interscapular BAT regions 78. Selective activation of ERα in female mice induces beiging of WAT and stimulates UCP-1 expression, acting protectively against metabolic dysfunction 113. In humans, decreased ESR1 and ESR2 mRNA levels have been reported in subcutaneous and visceral adipose tissue following bariatric surgery for the treatment of obesity, while ESR1 and ESR2 mRNA increased following surgery-induced weight loss 114. Conversely, others have shown that female mice lacking ERα expression in adipocytes are resistant to HFD-induced obesity and have improved glucose tolerance, suggesting a protective effect of reduced estrogen signalling in the context of metabolic dysfunction 115. These conflicting effects highlight the need to fully understand the effects of reduced estrogen signalling following menopause and the exact mechanisms underpinning changes in homeostasis and metabolic function.

Estrogens play an important role in the formation and maintenance of bone, which is variable across the lifespan. Estrogen receptors are expressed in osteoblasts (which promote bone formation), osteoclasts (which promote bone resorption) and osteocytes (mechanosensory cells) 116,117, allowing for estrogens to influence multiple aspects of bone homeostasis. Pre-menopause, estrogens act to limit osteoclast bone resorption and enhance bone formation by osteoblasts via the secretion of growth factors such as transforming growth factor-α 118. After menopause, falling estrogen levels are associated with changes in the expression of estrogen responsive genes, shifting bone homeostasis from formation to resorption by inducing the secretion of IL-6 and osteoclastogenesis. These changes reduce bone mineral density and increases both fracture rates and osteoporosis risk 119121. Many factors play a role in the rate of bone loss and increased resorption post menopause, including body weight and age at entering menopause, illustrating how the effects of estrogens on bone can interact with health and aging 29,122.

Muscle physiology is also influenced by circulating estrogens, with skeletal muscle expressing ERα in both mice and humans 123,124. ERα expression in muscle correlates with metabolic health in humans, in the context of female but not male physiology 125. Estrogens increase muscle strength and responses to anabolic exercise. Aside from the age-related decline in muscle strength that is irrespective of sex, the decline in estrogens during menopause specifically appears to reduce muscle function. In rodent models, OVX and muscle-specific elimination of ERα leads to reduced insulin sensitivity and mitochondrial function in skeletal muscle 126,127. Following MHT administration in humans, the data can be conflicting on its beneficial effects on muscle tissue. Isometric muscle strength, mobility and muscle power have all been reportedly improved, yet other studies have reported no improvements in skeletal muscle mass after estradiol implants 128,129.

Estrogen replacement can reverse homeostatic disruptions associated with menopause

The reduction of circulating estrogens during menopause is associated with a number of symptoms of disrupted homeostasis, such as hot flushes, cognitive impairment, vaginal atrophy, bone loss, sleep disturbances and mood alterations. Menopausal hormone therapy (MHT) is the standard treatment of care for the alleviation of these symptoms 101,130133. Multiple studies have shown that MHT containing estrogens is highly effective in improving hot flushes 134,135, vaginal atrophy 136, depression 137, metabolism 138, libido 139, sleep disturbances 140,141 and reducing osteoporosis risk 142,143 (Table 1).

Table 1 |.

MHT containing estrogens can alleviate a number of menopausal symptoms, such as sleep disturbances, vasomotor symptoms, mood disturbances, vaginal atrophy, osteoporosis, metabolic disruption and reduced libido

Menopausal Hormone Therapy (MHT) Non-Hormonal Treatment Options
Effective in mediating: Includes:
CNS derived symptoms including:

Sleep disturbances, hot flushes, mood alterations, brain fog
Neurokinin B Receptor Antagonists:

Fezolinetant - blocks neurokinin B receptor and KDNY neuron activity that stimulates thermoregulatory centres in hypothalamus, reducing hot flushes
Libido:

By acting on brain regions responsible for sexual function

Increases vaginal tone and blood flow
SERMs:

Ospemifene - reduces vaginal atrophy
Raloxifene - prevents osteoporosis

Vaginal Atrophy:

Maintains vaginal epithelium

Can improve sexual intercourse
SSRIs:

Depressive symptoms and anxiety, vasomotor symptoms (off-label)
Osteoporosis Risk:

Maintains bone density by limiting resorption
Protection against metabolic dysfunction:

Improves metabolism via mechanisms including mitochondrial protection

Data from refs. 129,180185. Non-hormonal options for menopausal symptoms include fezolinetant, a neurokinin B receptor antagonist, which reduces KNDY neuronal activity and reduces hot flushes186. Certain SERMs act agonistically or antagonistically on ERs in target tissues to improve symptoms, such as raloxifene on bone to improve osteoporosis and ospemifene on the vaginal epithelium to treat urinogenital symptoms. SSRIs, selective serotonin reuptake inhibitors.

Safety surrounding estrogens in MHT has been a contentious issue. Over 27,000 women were enrolled in the Women’s Health Initiative study, which aimed to ascertain if estrogens were protective against coronary heart disease. The estrogen plus progestin trial was stopped in 2002 due to reported increases in coronary events, invasive breast cancer, stroke and venous thromboembolism, while the estrogen only arm for those with a hysterectomy was ended in 2004 due to reported increased risks of stroke and breast cancer 144. Despite being one of the most prominent studies of MHT, there were many limitations including the advanced age of participants, a focus on relative rather than absolute risks and the predominantly white demographic of participants (Table 2). After initial findings were published, MHT use in the US fell by over 70% and reductions were also reported worldwide 145151.

Table 2 |.

Key MHT studies that have investigated the safety of hormone use for menopausal symptoms, the efficacy of unopposed versus combined therapies and the timing hypothesis of treatment initiation

Study Name Year Begun (data collection/recruitment) Type of Study Key research question Sample Size Hormone(s) used Key Conclusions Limitations
Women’s Health Initiative132, 187 1993 Long term observational clinical trial, community prevention study. Over 40 centres across the United States with additional satellite centres Evaluate HRT effects on CVD, cancer and osteoporosis. >27,000 Conjugated equine estrogen (CEE)+ medroxyprogesterone acetate

Or CEE only
Estrogen only: Increased risk in ischaemic stroke

Estrogen plus progestin: increased risk in coronary heart disease, breast cancer, reduced osteoporosis fractures and colorectal cancer132.
Predominantly white participants, study initiated in women who were recruited post menopause, emphasis of relative risk versus absolute risk188, 189
Heart and Estrogen/progestin Replacement Study190, 191 1994 Randomized, blinded, placebo-controlled secondary prevention trial. 20 centres throughout the United States Evaluate safety of estrogen plus progesterone against recurring heart disease. 2736 CEE+ medroxyprogesterone acetate

Or CEE only
No overall reduction in coronary heart disease with HRT in women with already established coronary heart disease. Predominantly white participants, >80% had minimum of high school education, only studied post- menopausal women.
Study of Women’s Health Across the Nation192 1994 Randomized, double-blind, placebo-controlled trial. 7 centres throughout United States Multi- ethnic study investigating physiological and psychological changes during menopause, beginning at the premenopausal stage. >3000 Premenopausal women, not taking hormones Transition to late perimenopause is when symptoms tend to be strongest (vasomotor symptoms, bone loss, depression, inflammation, sleep)15, 145, 193, 194. Despite being multi-ethnic, white participants were the biggest group, not all regions of US were represented ie south east192.
ELITE195 2004 Double blinded, placebo-controlled trial, recruitment largely based in California. Determine if HRT’s ability to reduce atherosclerosis is dependant on time of treatment initiation. 643 Oral 17βE2 only (hysterectomised people) or 17βE2 + vaginal micronized progesterone gel (uterus intact). When HRT initiated at time of menopause or <6 years after, CVD reduced. Supports Critical Therapeutic Window Hypothesis. Large proportion of participants were college graduates, largest proportion of participants were white.
KRONOS EARLY ESTROGEN PREVENTION STUDY196 2005 Randomised, double-blind, placebo-controlled trial, 9 centres across United States To investigate HRT effects on cardiovascular outcomes on recently post- menopausal women 727 Oral CEE or transdermal 17βE2. No increased risk reported on atherosclerosis progression after 4 years of either treatment. Central Europeans comprised majority of participants (81%)196.
Danish Osteoporosis Prevention Study (DOPS)197 1990 Prospective, investigator initiated trial, conducted at 4 centres in Denmark Investigate if HRT is effective as a primary preventative against osteoporotic fractures 2016 Oral 17βE2 + norethisterone acetate (intact) or oral 17βE2 (hysterectomy) When HRT was initiated early after menopause, mortality, heart failure and myocardial infarction was reduced, with no increase risk in stroke, cancer or venous thromboembolism198 Lack of placebo may have introduced bias199. Caucasian Danish participants only200.
THE ESTROGEN IN THE PREVENTION OF ATHEROSCLEROSIS TRIAL201 2001 Randomized, double blind, placebo-controlled, single centre, (California) carotid artery ultrasound trial Investigate the efficacy of unopposed estrogen to reduce atherosclerosis progression in post -menopausal women without CVD 222 Unopposed micronized 17βE2 or placebo. 17βE2 significantly reduced CVD. White women were most abundant (59%), >83% had greater than high school education.
ESTHER202 1999 Multi-centre (8 hospitals) case control study in France To investigate whether the route of administration of estrogen plays a role in VTE risk in people with a prothrombotic mutation Oral or transdermal estrogen Oral, but not transdermal estrogen, increases VTE risk. Only included postmenopausal women. Only included women in the vicinity of the hospitals ie from a small geographical area.
THE ESTROGEN REPLACEMENT AND ATHEROSCLEROSIS TRIAL203 2000 Randomized, double-blind, placebo-controlled clinical trial at 5 clinical sites in the United States Investigate the role of estrogen and/or estrogen plus progesterone on the progression of already developed coronary artery atherosclerosis in women 309 CEE only,

CEE+ medroxyprogesterone acetate,

or placebo
Neither treatment affected progression of the disease, therefore should not be used as a preventative if the disease has already been established. Age of women in study, questions surrounding relevance of cardiovascular endpoints, duration of menopause prior to ET or EPT commencement204.
Women's Angiographic Vitamin and Estrogen Trial205 2002 Randomised, double blind trial 7 clinical centres in the United States and Canada Investigate if estrogen or estrogen plus progestin therapy, either alone or in combination with antioxidant supplements, are effective in reducing 15–75% stenosis 423 CEE (hysterectomised participants) CEE+ medroxyprogesterone acetate (, intact participants)

or placebo.
Neither treatment displayed improvement of condition. Adherence to medications in the follow up portion varied between HRT assigned and vitamin assigned groups206.
ESTONIAN POSTMENOPAUSAL HORMONE THERAPY TRIAL207 1991 Blinded and unblinded HRT or placebo trial, 3 clinical centres in Estonia Investigate HRT on symptoms reported by women and quality of life 1823 CEE+ medroxyprogesterone acetate or placebo (blinded)

CEE+ medroxyprogesterone acetate

Placebo or no treatment (non-blinded)
Decreased sleeping problems and vasomotor symptoms with hormone therapy but increases in vaginal bleeding. No reported increase in quality of life. 28. Study centres were located in the two largest cities in Estonia, therefore rural populations may not have been represented208.

A meta-analysis of clinical trials with cardiovascular outcomes can be found in ref. 209. HRT, hormone replacement therapy; VTE, venous thromboembolism; ET, estrogen therapy; EPT, estrogen plus progestogen therapy.

The phase of menopause at which estrogen therapy is commenced is important, known as the critical therapeutic window hypothesis or timing hypothesis, originally proposed from pioneering work in atherosclerosis in aging female monkeys 152. If estrogens are given at or around the beginning of menopause, MHT can reduce the risk of cardiovascular disease 153. However, there is accumulating evidence that estrogenic MHT does not improve cardiovascular disease which has already been established in older post-menopausal research participants, as shown in the ELITE study, a randomized, double-blinded, placebo-controlled trial looking at atherosclerotic plaque progression during menopause 154. Similarly, estrogens can be protective against the progression of cognitive decline if started at or around menopause, whereas MHT does not seem to reduce already established cognitive impairment in later life 155.

It is also important to consider that MHT studies have historically been limited by the underrepresentation of many populations. The Women’s Health Initiative for example had predominantly white participants and in their estrogen plus progestin arm, less than 6.5% of women were black, 5.5% were Hispanic, 0.3% were Native American and 2.3% identified as Asian/Pacific Islander 156. Menopausal symptoms also vary among populations, with vasomotor symptoms reportedly more common 157 and more bothersome 158 in African American women than in white women.

MHT use is also disproportionate among groups. In the Carolina Breast Cancer Study, black women were less likely to use MHT overall but used unopposed estrogen formulations more than white women 159. Purported risks for breast cancer following MHT has also varied greatly between different populations 160162. Another limitation of several older MHT studies is the recent shift to using natural estrogens (such as estradiol) and progestogens (such as progesterone), along with other estrogen treatments that bypass the first pass effect of oral administration by the liver, including vaginal patches and creams. Earlier studies assessing risk factors of MHT use with cardiovascular events often used conjugated equine estrogens (CEEs) like Premarin or progestins (synthetic progestogens), whereas a recent review has suggested that estradiol may have a reduced risk in initiating cardiovascular disease in comparison with CEEs and act more protectively against bone fracture or cognitive decline 163.

Non-hormonal alternatives to MHT

The approval of fezolinetant by the FDA has been the most recent non-hormonal treatment for menopause, effective in reducing moderate to severe vasomotor symptoms 164. When estrogens in the circulation are low, GnRH neurons are stimulated to release LH in a pulsatile fashion, which is often correlated with a rise in skin temperature. This release of LH is thought to be mediated by kisspeptin expressing neurons, which also co-express neurokinin B in the hypothalamus 165,166. Kisspeptin, neurokinin B and dynorphin (KNDY) neurons within the hypothalamus modulate hot flush activity 167. Therefore Fezolinetant, a highly selective treatment that antogonizes the neurokinin B receptor, has emerged as an effective treatment for vasomotor symptoms resulting from low levels of circulating estrogens during menopause. Other treatments include selective serotonin reuptake inhibitors (SSRIs) for the treatment depressive symptoms, however they are generally not effective for weight gain or severe vasomotor symptoms 168. Phytoestrogens are a compound found in plants which have similar properties to estradiol 169. While some studies have shown their efficacy against hot flushes, there have also been reports of altered HPG axis signalling and adverse effects on reproductive organs 169,170.

Selective estrogen receptor modulators (SERMS) are synthetic, nonsteroidal ligands that can act either as estrogen receptor agonists or antagonists depending on the target gene or tissue, where they compete against estrogens and cause confirmational changes to ligand-receptor structural complexes 171. SERMS currently approved by the FDA include Tamoxifen, Raloxifene and Bazedoxifene. A significant limitation of these ligands however, is the lack of an ideal SERM which acts agonistically in tissues where estrogenic action is desirable, such as muscle, bone or even the brain, but inhibits estrogenic action in reproductive tissues 172. Tamoxifen is a commonly used SERM to treat invasive breast cancer which functions as an antagonist and agonist in the breast and uterus respectively, with the latter increasing the risk of endometrial hyperplasia 173. Tamoxifen also causes hot flushes in over 70% of patients, but the mechanism is poorly understood 174. Raloxifene is a SERM prescribed to treat osteoporosis due to its agonistic actions in bone, but can also increase the risk of deep vein thrombosis or further exacerbate stroke risk in patients with an already elevated cardiovascular disease risk profile. Raloxifene also is not effective for treating hot flushes and may also exasperate vasomotor symptoms. 175177.

There are currently no SERMS approved for the treatment of hot flushes, however bazedoxifene can be useful when prescribed in conjunction with a conjugated equine estrogen in what is known as a tissue selective estrogen complex 178. This is generally suitable when the uterus is intact and it is greater than a year since the last menstrual period. The clinical benefit of using tissue selective estrogen complexes over traditional MHT include the prevention of bone loss while avoiding proliferation of breast or endometrium tissue due its estrogen antagonistic properties, removing the need for a progestogen in combination with a conjugated equine estrogen 179. Ospemifene, a SERM similar in structure to tamoxifen, was approved by the FDA in the last decade for the treatment of vulvovaginal atrophy and acts agonistically in vaginal epithelium, with minimal effects on the endometrium 180. However, clinical trials involving ospemifene ran for only 52 weeks, therefore the longer acting effects of this SERM are unknown 181.

Opportunities and the future for targeting estrogen receptors in MHT

The past two decades have seen an increased application of genetic manipulations to understand the roles of specific estrogen receptor signalling pathways in different tissues, cell types and neuronal populations, predominantly using rodents as a model. This has revealed profound impacts of estrogen receptor signalling at specific neurons on whole body energy expenditure and homeostasis, and at the same time important local actions of estrogens in tissues like liver and fat that also play key roles in energy metabolism and storage. Despite this knowledge, estrogen-related hormone treatments for menopause have barely progressed over the same period, with the only major new treatment for menopause symptoms targeting kisspeptin expressing neurons, that likely manifest only a subset of physiological changes that occur with the menopause transition. The development of treatments that can ameliorate the constellation of homeostatic changes, without stimulating tissues that might increase cancer or cardiovascular disease risk, could provide major improvements in menopausal care.

One strategy to specifically target estrogens is the use of estrogen associated prodrugs that are metabolized to produce bioactive estrogens in certain tissues, while reducing off target effects. Given the role of the brain in mediating many of the effects of estrogens in health and homeostasis, prodrugs have been created that target bioactive estrogens specifically to central nervous system (CNS). Currently under investigation is the novel prodrug of 17βE2, 10β,17β-dihydroxyestra-1,4-dien-3-one (DHED). DHED is an inert derivative of 17βE2 which is enzymatically converted to estradiol exclusively within the brain by a CNS specific enzyme and has a short acting half-life, leading to brain-specific estrogenic activity without actions in the periphery. DHED has already improved symptoms in animal models of CNS regulated conditions such as cognitive decline, stroke, hot flush and depression 182,183. Whether DHED has the potential to exert protective effects against symptoms originating in the periphery such as bone loss remains to be investigated. Therefore, the generation of prodrugs or other pharmacological agents which selectively target estrogens or their signalling pathways may pave the way for effective alternative hormonal treatments, while reducing off- target effects.

Treatments that stimulate or inhibit specific pathways downstream of estrogen receptors may also be an alternative option to broaden therapeutic options with estrogens. For example, pathway preferential estrogens (often referred to as PaPes) have been developed that have a lower binding affinity for ERα than 17βE2. Consequently, binding of pathway preferential estrogens is insufficient to sustain nuclear-pathway activity of ERα, but is still effective to stimulate the extranuclear pathway. Pathway preferential estrogens have been shown to reduce adiposity, body weight and triglyceride levels in mice, without causing proliferation of mammary, thymus or uterine tissue, as well as exerting protective effects on cardiovascular epithelium in OVX mice 184,185.

Another possibility is the incorporation of other estrogens currently not widely used in MHT. 17αE2 has already been discussed as a lifespan extending drug preferentially favouring male mice 51,54. Continuing to understand the sexually dimorphic patterns underpinning the signalling of this estrogen could help uncover male-specific mechanisms which have the potential to be beneficially targeted during female aging. Estetrol (E4) produced by the fetus during pregnancy is thought to have more tissue-specific and favourable actions on organs such as the liver 186. E4 has also been shown to improve vasomotor symptoms in rodent hot flush models 187, was promising in a phase I clinical trial, and is currently under further development for use against hot flushes 188. Estriol (E3) also has a role for reducing menopausal symptoms, however a number of studies are now quite dated. E3 is metabolised from estradiol and has been shown to improve climactic symptoms while not inducing proliferation of the endometrium or breast tissue in a Japanese cohort 189. Transvaginal estriol has also proven effective in treating vaginal atrophy 190. Estriol has also been shown as a safe method of MHT in a phase III clinical trial in those with ER positive breast cancer 191. The more widespread incorporation of alternative estrogens to those commonly used may therefore be at the forefront of future MHT treatments.

Conclusions

Estrogen signalling has widespread and coordinated effects across the brain and body to maintain energy homeostasis. In addition to preserving energy balance, ovarian estrogens have the potential to alter lifespan, either directly or indirectly. These intercalated effects of estrogens are disrupted during aging, which is associated with several metabolic perturbations and increased mortality. While MHT can alleviate some of these symptoms and provide overall benefit, studies involving MHT risk profiles have had severe limitations and future studies encompassing more appropriate groups are needed before conclusive thoughts on estrogen use at midlife and through aging can be critically assessed. Newer non-hormonal alternatives have the exciting potential to provide more nuanced therapeutic options, targeting tissues or pathways with varying levels of selectivity or specificity. A move toward more precise therapies may limit the benefits to certain tissues or side effects, but also provide an opportunity to minimize risks. Ultimately, understanding the effects of estrogens across many tissues and life stages will be foundational for counteracting the suite of homeostatic disruptions associated with menopause.

Acknowledgements

The authors thank scholarships to CC from the Elman Poole Trust, The Hope Foundation of New Zealand, The Collaboration of Ageing Research Excellence at the University of Otago and the Australasian Menopause Society. SMC was supported by NIH grants AG066821 and DK136073.

Footnotes

The authors declare no competing interests.

References:

  • 1.Dillin A, Gottschling DE, Nyström T. The good and the bad of being connected: the integrons of aging. Curr Opin Cell Biol. 2014;26:107–112. doi: 10.1016/j.ceb.2013.12.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Pataky MW, Young WF, Nair KS. Hormonal and Metabolic Changes of Aging and the Influence of Lifestyle Modifications. Mayo Clin Proc. 2021;96(3):788–814. doi: 10.1016/j.mayocp.2020.07.033 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Roh E, Song DK, Kim MS. Emerging role of the brain in the homeostatic regulation of energy and glucose metabolism. Exp Mol Med. 2016;48(3):e216–e216. doi: 10.1038/emm.2016.4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Marsh ML, Oliveira MN, Vieira-Potter VJ. Adipocyte Metabolism and Health after the Menopause: The Role of Exercise. Nutrients. 2023;15(2):444. doi: 10.3390/nu15020444 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Stepniak J, Karbownik-Lewinska M. 17β-estradiol prevents experimentally-induced oxidative damage to membrane lipids and nuclear DNA in porcine ovary. Syst Biol Reprod Med. 2016;62(1):17–21. doi: 10.3109/19396368.2015.1101510 [DOI] [PubMed] [Google Scholar]
  • 6.Malespin M, Nassri A. Endocrine Diseases and the Liver: An Update. Clin Liver Dis. 2019;23(2):233–246. doi: 10.1016/j.cld.2018.12.006 [DOI] [PubMed] [Google Scholar]
  • 7.Hess RA. Estrogen in the adult male reproductive tract: A review. Reprod Biol Endocrinol RBE. 2003;1:52. doi: 10.1186/1477-7827-1-52 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lee HR, Kim TH, Choi KC. Functions and physiological roles of two types of estrogen receptors, ERα and ERβ, identified by estrogen receptor knockout mouse. Lab Anim Res. 2012;28(2):71–76. doi: 10.5625/lar.2012.28.2.71 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.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. doi: 10.1530/JME-15-0254 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Tang ZR, Zhang R, Lian ZX, Deng SL, Yu K. Estrogen-Receptor Expression and Function in Female Reproductive Disease. Cells. 2019;8(10):1123. doi: 10.3390/cells8101123 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Osterlund MK, Keller E, Hurd YL. The human forebrain has discrete estrogen receptor alpha messenger RNA expression: high levels in the amygdaloid complex. Neuroscience. 2000;95(2):333–342. doi: 10.1016/s0306-4522(99)00443-1 [DOI] [PubMed] [Google Scholar]
  • 12.Wilson ME, Westberry JM, Prewitt AK. Dynamic Regulation of Estrogen Receptor-Alpha Gene Expression in the Brain: A Role for Promoter Methylation? Front Neuroendocrinol. 2008;29(3):375–385. doi: 10.1016/j.yfrne.2008.03.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Liu X, Shi H. Regulation of Estrogen Receptor α Expression in the Hypothalamus by Sex Steroids: Implication in the Regulation of Energy Homeostasis. Int J Endocrinol. 2015;2015:949085. doi: 10.1155/2015/949085 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Shughrue PJ, Komm B, Merchenthaler I. The distribution of estrogen receptor-beta mRNA in the rat hypothalamus. Steroids. 1996;61(12):678–681. doi: 10.1016/s0039-128x(96)00222-x [DOI] [PubMed] [Google Scholar]
  • 15.Björnström L, Sjöberg M. Mechanisms of Estrogen Receptor Signaling: Convergence of Genomic and Nongenomic Actions on Target Genes. Mol Endocrinol. 2005;19(4):833–842. doi: 10.1210/me.2004-0486 [DOI] [PubMed] [Google Scholar]
  • 16.Fuentes N, Silveyra P. Estrogen receptor signaling mechanisms. Adv Protein Chem Struct Biol. 2019;116:135–170. doi: 10.1016/bs.apcsb.2019.01.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Marino M, Galluzzo P, Ascenzi P. Estrogen signaling multiple pathways to impact gene transcription. Curr Genomics. 2006;7(8):497–508. doi: 10.2174/138920206779315737 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Méndez-Reséndiz KA, Enciso-Pablo Ó, González-Ramírez R, Juárez-Contreras R, Rosenbaum T, Morales-Lázaro SL. Steroids and TRP Channels: A Close Relationship. Int J Mol Sci. 2020;21(11):3819. doi: 10.3390/ijms21113819 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Burger HG, Hale GE, Dennerstein L, Robertson DM. Cycle and hormone changes during perimenopause: the key role of ovarian function. Menopause. 2008;15(4):603. doi: 10.1097/gme.0b013e318174ea4d [DOI] [PubMed] [Google Scholar]
  • 20.Wang X, Wang L, Xiang W. Mechanisms of ovarian aging in women: a review. J Ovarian Res. 2023;16:67. doi: 10.1186/s13048-023-01151-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Reyes FI, Winter JS, Faiman C. Pituitary-ovarian relationships preceding the menopause. I. A cross-sectional study of serum follice-stimulating hormone, luteinizing hormone, prolactin, estradiol, and progesterone levels. Am J Obstet Gynecol. 1977;129(5):557–564. [PubMed] [Google Scholar]
  • 22.Longcope C, Franz C, Morello C, Baker R, Johnston CC. Steroid and gonadotropin levels in women during the peri-menopausal years. Maturitas. 1986;8(3):189–196. doi: 10.1016/0378-5122(86)90025-3 [DOI] [PubMed] [Google Scholar]
  • 23.Al-Azzawi F, Palacios S. Hormonal changes during menopause. Maturitas. 2009;63(2):135–137. doi: 10.1016/j.maturitas.2009.03.009 [DOI] [PubMed] [Google Scholar]
  • 24.Ambikairajah A, Walsh E, Tabatabaei-Jafari H, Cherbuin N. Fat mass changes during menopause: a metaanalysis. Am J Obstet Gynecol. 2019;221(5):393–409.e50. doi: 10.1016/j.ajog.2019.04.023 [DOI] [PubMed] [Google Scholar]
  • 25.Greendale GA, Sternfeld B, Huang M, et al. Changes in body composition and weight during the menopause transition. JCI Insight. 2019;4(5). doi: 10.1172/jci.insight.124865 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Gordon T, Kannel WB, Hjortland MC, McNamara PM. Menopause and coronary heart disease. The Framingham Study. Ann Intern Med. 1978;89(2):157–161. doi: 10.7326/0003-4819-89-2-157 [DOI] [PubMed] [Google Scholar]
  • 27.Mendelsohn ME. Protective effects of estrogen on the cardiovascular system. Am J Cardiol. 2002;89(12, Supplement 1):12–17. doi: 10.1016/S0002-9149(02)02405-0 [DOI] [PubMed] [Google Scholar]
  • 28.Recker R, Lappe J, Davies K, Heaney R. Characterization of perimenopausal bone loss: a prospective study. J Bone Miner Res Off J Am Soc Bone Miner Res. 2000;15(10):1965–1973. doi: 10.1359/jbmr.2000.15.10.1965 [DOI] [PubMed] [Google Scholar]
  • 29.Finkelstein JS, Brockwell SE, Mehta V, et al. Bone mineral density changes during the menopause transition in a multiethnic cohort of women. J Clin Endocrinol Metab. 2008;93(3):861–868. doi: 10.1210/jc.2007-1876 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Anagnostis P, Siolos P, Gkekas NK, et al. Association between age at menopause and fracture risk: a systematic review and meta-analysis. Endocrine. 2019;63(2):213–224. doi: 10.1007/s12020-018-1746-6 [DOI] [PubMed] [Google Scholar]
  • 31.Dobbs MB, Buckwalter J, Saltzman C. Osteoporosis. Iowa Orthop J. 1999;19:43–52. [PMC free article] [PubMed] [Google Scholar]
  • 32.Sun L, Peng Y, Sharrow AC, et al. FSH directly regulates bone mass. Cell. 2006;125(2):247–260. doi: 10.1016/j.cell.2006.01.051 [DOI] [PubMed] [Google Scholar]
  • 33.Rocca WA, Grossardt BR, de Andrade M, Malkasian GD, Melton LJ. Survival patterns after oophorectomy in premenopausal women: a population-based cohort study. Lancet Oncol. 2006;7(10):821–828. doi: 10.1016/S1470-2045(06)70869-5 [DOI] [PubMed] [Google Scholar]
  • 34.Parker WH, Feskanich D, Broder MS, et al. Long-term mortality associated with oophorectomy compared with ovarian conservation in the nurses’ health study. Obstet Gynecol. 2013;121(4):709–716. doi: 10.1097/AOG.0b013e3182864350 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Gottschau M, Rosthøj S, Settnes A, et al. Long-Term Health Consequences After Ovarian Removal at Benign Hysterectomy : A Nationwide Cohort Study. Ann Intern Med. 2023;176(5):596–604. doi: 10.7326/M22-1628 [DOI] [PubMed] [Google Scholar]
  • 36.Melton LJ, Crowson CS, Malkasian GD, O’Fallon WM. Fracture risk following bilateral oophorectomy. J Clin Epidemiol. 1996;49(10):1111–1115. doi: 10.1016/0895-4356(96)00211-9 [DOI] [PubMed] [Google Scholar]
  • 37.Rocca WA, Shuster LT, Grossardt BR, et al. Long-Term Effects of Bilateral Oophorectomy on Brain Aging: Unanswered Questions from the Mayo Clinic Cohort Study of Oophorectomy and Aging. Womens Health. 2009;5(1):39–48. doi: 10.2217/17455057.5.1.39 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Rocca WA, Grossardt BR, Geda YE, et al. Long-term risk of depressive and anxiety symptoms after early bilateral oophorectomy. Menopause N Y N. 2008;15(6):1050–1059. doi: 10.1097/gme.0b013e318174f155 [DOI] [PubMed] [Google Scholar]
  • 39.Jacoby VL, Grady D, Wactawski-Wende J, et al. Oophorectomy vs ovarian conservation with hysterectomy: cardiovascular disease, hip fracture, and cancer in the Women’s Health Initiative Observational Study. Arch Intern Med. 2011;171(8):760–768. doi: 10.1001/archinternmed.2011.121 [DOI] [PubMed] [Google Scholar]
  • 40.Finch A, Metcalfe KA, Chiang JK, et al. The impact of prophylactic salpingo-oophorectomy on menopausal symptoms and sexual function in women who carry a BRCA mutation. Gynecol Oncol. 2011;121(1):163–168. doi: 10.1016/j.ygyno.2010.12.326 [DOI] [PubMed] [Google Scholar]
  • 41.Atsma F, Bartelink MLEL, Grobbee DE, van der Schouw YT. Postmenopausal status and early menopause as independent risk factors for cardiovascular disease: a meta-analysis. Menopause N Y N. 2006;13(2):265–279. doi: 10.1097/01.gme.0000218683.97338.ea [DOI] [PubMed] [Google Scholar]
  • 42.Mueller K, Hsiao S. Estrus- and ovariectomy-induced body weight changes: evidence for two estrogenic mechanisms. J Comp Physiol Psychol. 1980;94(6):1126–1134. doi: 10.1037/h0077746 [DOI] [PubMed] [Google Scholar]
  • 43.Rogers NH, Perfield JW II, Strissel KJ, Obin MS, Greenberg AS. Reduced Energy Expenditure and Increased Inflammation Are Early Events in the Development of Ovariectomy-Induced Obesity. Endocrinology. 2009;150(5):2161–2168. doi: 10.1210/en.2008-1405 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Stubbins RE, Holcomb VB, Hong J, Núñez NP. Estrogen modulates abdominal adiposity and protects female mice from obesity and impaired glucose tolerance. Eur J Nutr. 2012;51(7):861–870. doi: 10.1007/s00394-011-0266-4 [DOI] [PubMed] [Google Scholar]
  • 45.Wang Y, Shoemaker R, Thatcher SE, Batifoulier-Yiannikouris F, English VL, Cassis LA. Administration of 17β-estradiol to ovariectomized obese female mice reverses obesity-hypertension through an ACE2-dependent mechanism. Am J Physiol - Endocrinol Metab. 2015;308(12):E1066–E1075. doi: 10.1152/ajpendo.00030.2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Cintron D, Beckman JP, Bailey KR, Lahr BD, Jayachandran M, Miller VM. Plasma orexin A levels in recently menopausal women during and 3 years following use of hormone therapy. Maturitas. 2017;99:59–65. doi: 10.1016/j.maturitas.2017.01.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Asdell SA, Joshi SR. Reproduction and Longevity in the Hamster and Rat. Biol Reprod. 1976;14(4):478–480. doi: 10.1095/biolreprod14.4.478 [DOI] [PubMed] [Google Scholar]
  • 48.Phelan JP. Reproductive Costs and Longevity in the House Mouse. Ph.D. Harvard University; 1995. Accessed March 21, 2024. https://www.proquest.com/docview/304175366/abstract/E30CD7AD9318428DPQ/1 [Google Scholar]
  • 49.Mühlbock O Factors Influencing the Life-Span of Inbred Mice. Gerontologia. 2009;3(4):177–183. doi: 10.1159/000210897 [DOI] [PubMed] [Google Scholar]
  • 50.Arriola Apelo SI, Lin A, Brinkman JA, et al. Ovariectomy uncouples lifespan from metabolic health and reveals a sex-hormone-dependent role of hepatic mTORC2 in aging. Galvan V, Tyler JK, eds. eLife. 2020;9:e56177. doi: 10.7554/eLife.56177 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Strong R, Miller RA, Antebi A, et al. Longer lifespan in male mice treated with a weakly estrogenic agonist, an antioxidant, an α-glucosidase inhibitor or a Nrf2-inducer. Aging Cell. 2016;15(5):872–884. doi: 10.1111/acel.12496 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Harrison DE, Strong R, Reifsnyder P, et al. 17-a-estradiol late in life extends lifespan in aging UM-HET3 male mice; nicotinamide riboside and three other drugs do not affect lifespan in either sex. Aging Cell. 2021;20(5):e13328. doi: 10.1111/acel.13328 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Garratt M, Bower B, Garcia GG, Miller RA. Sex differences in lifespan extension with acarbose and 17-α estradiol: gonadal hormones underlie male-specific improvements in glucose tolerance and mTORC2 signaling. Aging Cell. 2017;16(6):1256–1266. doi: 10.1111/acel.12656 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Stout MB, Steyn FJ, Jurczak MJ, et al. 17α-Estradiol Alleviates Age-related Metabolic and Inflammatory Dysfunction in Male Mice Without Inducing Feminization. J Gerontol A Biol Sci Med Sci. 2017;72(1):3–15. doi: 10.1093/gerona/glv309 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Camon C, Prescott M, Neyt C, et al. Systemic metabolic benefits of 17α-estradiol are not exclusively mediated by ERα in glutamatergic or GABAergic neurons. GeroScience. Published online May 22, 2024. doi: 10.1007/s11357-024-01192-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Garratt M, Leander D, Pifer K, et al. 17-α estradiol ameliorates age-associated sarcopenia and improves late-life physical function in male mice but not in females or castrated males. Aging Cell. 2019;18(2):e12920. doi: 10.1111/acel.12920 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Mann SN, Hadad N, Nelson Holte M, et al. Health benefits attributed to 17α-estradiol, a lifespan-extending compound, are mediated through estrogen receptor α. eLife. 2020;9:e59616. doi: 10.7554/eLife.59616 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Steyn FJ, Ngo ST, Chen VP, et al. 17α-estradiol acts through hypothalamic pro-opiomelanocortin expressing neurons to reduce feeding behavior. Aging Cell. 2018;17(1):e12703. doi: 10.1111/acel.12703 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Regan JC, Partridge L. Gender and longevity: why do men die earlier than women? Comparative and experimental evidence. Best Pract Res Clin Endocrinol Metab. 2013;27(4):467–479. doi: 10.1016/j.beem.2013.05.016 [DOI] [PubMed] [Google Scholar]
  • 60.Austad SN, Fischer KE. Sex Differences in Lifespan. Cell Metab. 2016;23(6):1022–1033. doi: 10.1016/j.cmet.2016.05.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Miller RA, Harrison DE, Cortopassi GA, et al. Lifespan effects in male UM-HET3 mice treated with sodium thiosulfate, 16-hydroxyestriol, and late-start canagliflozin. GeroScience. Published online May 16, 2024. doi: 10.1007/s11357-024-01176-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Cargill SL, Carey JR, Müller HG, Anderson G. Age of ovary determines remaining life expectancy in old ovariectomized mice. Aging Cell. 2003;2(3):185–190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Mason JB, Cargill SL, Anderson GB, Carey JR. Transplantation of Young Ovaries to Old Mice Increased Life Span in Transplant Recipients. J Gerontol A Biol Sci Med Sci. 2009;64A(12):1207–1211. doi: 10.1093/gerona/glp134 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Mason JB, Cargill SL, Griffey SM, Reader JR, Anderson GB, Carey JR. Transplantation of Young Ovaries Restored Cardioprotective Influence in Postreproductive-Aged Mice. Aging Cell. 2011;10(3):448–456. doi: 10.1111/j.1474-9726.2011.00691.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Habermehl TL, Parkinson KC, Hubbard GB, et al. Extension of longevity and reduction of inflammation is ovarian-dependent, but germ cell-independent in post-reproductive female mice. GeroScience. 2019;41(1):25–38. doi: 10.1007/s11357-018-0049-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Buffenstein R, Poppitt SD, McDevitt RM, Prentice AM. Food intake and the menstrual cycle: a retrospective analysis, with implications for appetite research. Physiol Behav. 1995;58(6):1067–1077. doi: 10.1016/0031-9384(95)02003-9 [DOI] [PubMed] [Google Scholar]
  • 67.Fessler DMT. No time to eat: an adaptationist account of periovulatory behavioral changes. Q Rev Biol. 2003;78(1):3–21. doi: 10.1086/367579 [DOI] [PubMed] [Google Scholar]
  • 68.Slonaker JR. The effect of pubescence, oestruation and menopause on the voluntary activity in the albino rat. Am J Physiol-Leg Content. 1924;68(2):294–315. doi: 10.1152/ajplegacy.1924.68.2.294 [DOI] [Google Scholar]
  • 69.Brobeck JR, Wheatland M, Strominger JL. Variations in regulation of energy exchange associated with estrus, diestrus and pseudopregnancy in rats. Endocrinology. 1947;40(2):65–72. doi: 10.1210/endo-40-2-65 [DOI] [PubMed] [Google Scholar]
  • 70.Kopp C, Ressel V, Wigger E, Tobler I. Influence of estrus cycle and ageing on activity patterns in two inbred mouse strains. Behav Brain Res. 2006;167(1):165–174. doi: 10.1016/j.bbr.2005.09.001 [DOI] [PubMed] [Google Scholar]
  • 71.Olofsson LE, Pierce AA, Xu AW. Functional requirement of AgRP and NPY neurons in ovarian cycle-dependent regulation of food intake. Proc Natl Acad Sci U S A. 2009;106(37):15932–15937. doi: 10.1073/pnas.0904747106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Sanchez-Alavez M, Alboni S, Conti B. Sex- and age-specific differences in core body temperature of C57Bl/6 mice. Age. 2011;33(1):89–99. doi: 10.1007/s11357-010-9164-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Heine PA, Taylor JA, Iwamoto GA, Lubahn DB, Cooke PS. Increased adipose tissue in male and female estrogen receptor-α knockout mice. Proc Natl Acad Sci. 2000;97(23):12729–12734. doi: 10.1073/pnas.97.23.12729 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Park S, Zhao Y, Yoon S, et al. Repressor of Estrogen Receptor Activity (REA) Is Essential for Mammary Gland Morphogenesis and Functional Activities: Studies in Conditional Knockout Mice. Endocrinology. 2011;152(11):4336–4349. doi: 10.1210/en.2011-1100 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Xu Y, Nedungadi TP, Zhu L, et al. Distinct Hypothalamic Neurons Mediate Estrogenic Effects on Energy Homeostasis and Reproduction. Cell Metab. 2011;14(4):453–465. doi: 10.1016/j.cmet.2011.08.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Geary N, Asarian L, Korach KS, Pfaff DW, Ogawa S. Deficits in E2-Dependent Control of Feeding, Weight Gain, and Cholecystokinin Satiation in ER-α Null Mice. Endocrinology. 2001;142(11):4751–4757. doi: 10.1210/endo.142.11.8504 [DOI] [PubMed] [Google Scholar]
  • 77.Roepke TA, Xue C, Bosch MA, Scanlan TS, Kelly MJ, Rønnekleiv OK. Genes associated with membrane-initiated signaling of estrogen and energy homeostasis. Endocrinology. 2008;149(12):6113–6124. doi: 10.1210/en.2008-0769 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Martínez de Morentin PB, González-García I, Martins L, et al. Estradiol Regulates Brown Adipose Tissue Thermogenesis via Hypothalamic AMPK. Cell Metab. 2014;20(1):41–53. doi: 10.1016/j.cmet.2014.03.031 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Correa SM, Newstrom DW, Warne JP, et al. An estrogen-responsive module in the ventromedial hypothalamus selectively drives sex-specific activity in females. Cell Rep. 2015;10(1):62–74. doi: 10.1016/j.celrep.2014.12.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.van Veen JE, Kammel LG, Bunda PC, et al. Hypothalamic estrogen receptor alpha establishes a sexually dimorphic regulatory node of energy expenditure. Nat Metab. 2020;2(4):351–363. doi: 10.1038/s42255-020-0189-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Krause WC, Rodriguez R, Gegenhuber B, et al. Estrogen Drives Brain Melanocortin to Increase Physical Activity in Females. Nature. 2021;599(7883):131–135. doi: 10.1038/s41586-021-04010-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Spiteri T, Ogawa S, Musatov S, Pfaff DW, Agmo A. The role of the estrogen receptor α in the medial preoptic area in sexual incentive motivation, proceptivity and receptivity, anxiety, and wheel running in female rats. Behav Brain Res. 2012;230(1):11–20. doi: 10.1016/j.bbr.2012.01.048 [DOI] [PubMed] [Google Scholar]
  • 83.Santollo J, Daniels D. Anorexigenic effects of estradiol in the medial preoptic area occur through membrane-associated estrogen receptors and metabotropic glutamate receptors. Horm Behav. 2019;107:20–25. doi: 10.1016/j.yhbeh.2018.11.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Puelles L Survey of Midbrain, Diencephalon, and Hypothalamus Neuroanatomic Terms Whose Prosomeric Definition Conflicts With Columnar Tradition. Front Neuroanat. 2019;13:20. doi: 10.3389/fnana.2019.00020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Xu P, Cao X, He Y, et al. Estrogen receptor–α in medial amygdala neurons regulates body weight. J Clin Invest. 2015;125(7):2861–2876. doi: 10.1172/JCI80941 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Wilson ME, Rosewell KL, Kashon ML, Shughrue PJ, Merchenthaler I, Wise PM. Age differentially influences estrogen receptor-α (ERα) and estrogen receptor-β (ERβ) gene expression in specific regions of the rat brain. Mech Ageing Dev. 2002;123(6):593–601. doi: 10.1016/S0047-6374(01)00406-7 [DOI] [PubMed] [Google Scholar]
  • 87.Thakur MK, Sharma PK. Aging of Brain: Role of Estrogen. Neurochem Res. 2006;31(11):1389–1398. doi: 10.1007/s11064-006-9191-y [DOI] [PubMed] [Google Scholar]
  • 88.Foster TC, Sharrow KM, Kumar A, Masse J. Interaction of age and chronic estradiol replacement on memory and markers of brain aging. Neurobiol Aging. 2003;24(6):839–852. doi: 10.1016/S0197-4580(03)00014-9 [DOI] [PubMed] [Google Scholar]
  • 89.Hajdarovic KH, Yu D, Hassell LA, et al. Single-cell analysis of the aging female mouse hypothalamus. Nat Aging. 2022;2(7):662–678. doi: 10.1038/s43587-022-00246-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Sharma G, Prossnitz ER. Targeting the G protein-coupled estrogen receptor (GPER) in obesity and diabetes. Endocr Metab Sci. 2021;2:100080. doi: 10.1016/j.endmts.2021.100080 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Davis KE, Carstens EJ, Irani BG, Gent LM, Hahner LM, Clegg DJ. Sexually dimorphic role of G protein-coupled estrogen receptor (GPER) in modulating energy homeostasis. Horm Behav. 2014;66(1):196–207. doi: 10.1016/j.yhbeh.2014.02.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Della Torre S, Rando G, Meda C, et al. Amino Acid-Dependent Activation of Liver Estrogen Receptor Alpha Integrates Metabolic and Reproductive Functions via IGF-1. Cell Metab. 2011;13(2):205–214. doi: 10.1016/j.cmet.2011.01.002 [DOI] [PubMed] [Google Scholar]
  • 93.Mondal SA, Mann SN, van der Linden C, et al. Metabolic benefits of 17α-estradiol in liver are partially mediated by ERβ in male mice. BioRxiv Prepr Serv Biol. Published online March 25, 2023:2023.03.25.534216. doi: 10.1101/2023.03.25.534216 [DOI] [Google Scholar]
  • 94.Qiu S, Vazquez JT, Boulger E, et al. Hepatic estrogen receptor α is critical for regulation of gluconeogenesis and lipid metabolism in males. Sci Rep. 2017;7(1):1661. doi: 10.1038/s41598-017-01937-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Dahlman I, Vaxillaire M, Nilsson M, et al. Estrogen receptor alpha gene variants associate with type 2 diabetes and fasting plasma glucose. Pharmacogenet Genomics. 2008;18(11):967. doi: 10.1097/FPC.0b013e32831101ef [DOI] [PubMed] [Google Scholar]
  • 96.Chow JDY, Jones MEE, Prelle K, Simpson ER, Boon WC. A selective estrogen receptor α agonist ameliorates hepatic steatosis in the male aromatase knockout mouse. J Endocrinol. 2011;210(3):323–334. doi: 10.1530/JOE-10-0462 [DOI] [PubMed] [Google Scholar]
  • 97.Della Torre S, Mitro N, Fontana R, et al. An Essential Role for Liver ERα in Coupling Hepatic Metabolism to the Reproductive Cycle. Cell Rep. 2016;15(2):360–371. doi: 10.1016/j.celrep.2016.03.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Francavilla A, Di Leo A, Eagon PK, et al. Regenerating Rat Liver: Correlations Between Estrogen Receptor Localization and Deoxyribonucleic Acid Synthesis. Gastroenterology. 1984;86(3):552–557. [PMC free article] [PubMed] [Google Scholar]
  • 99.Srisowanna N, Choijookhuu N, Yano K, et al. The Effect of Estrogen on Hepatic Fat Accumulation during Early Phase of Liver Regeneration after Partial Hepatectomy in Rats. Acta Histochem Cytochem. 2019;52(4):67–75. doi: 10.1267/ahc.19018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Kim SE, Min JS, Lee S, Lee DY, Choi D. Different effects of menopausal hormone therapy on non-alcoholic fatty liver disease based on the route of estrogen administration. Sci Rep. 2023;13(1):15461. doi: 10.1038/s41598-023-42788-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Mattsson LA, Skouby S, Rees M, et al. Efficacy and tolerability of continuous combined hormone replacement therapy in early postmenopausal women. Menopause Int. 2007;13(3):124–131. doi: 10.1258/175404507781605596 [DOI] [PubMed] [Google Scholar]
  • 102.Wang Y, Wang Y, Liu L, Cui H. Ovariectomy induces abdominal fat accumulation by improving gonadotropin-releasing hormone secretion in mouse. Biochem Biophys Res Commun. 2022;588:111–117. doi: 10.1016/j.bbrc.2021.12.039 [DOI] [PubMed] [Google Scholar]
  • 103.Nishio E, Hayashi T, Nakatani M, et al. Lack of association of ovariectomy-induced obesity with overeating and the reduction of physical activities. Biochem Biophys Rep. 2019;20:100671. doi: 10.1016/j.bbrep.2019.100671 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Camara C, Zhou L yuan, Ma Y, et al. Effect of ovariectomy on serum adiponectin levels and visceral fat in rats. J Huazhong Univ Sci Technolog Med Sci. 2014;34(6):825–829. doi: 10.1007/s11596-014-1360-7 [DOI] [PubMed] [Google Scholar]
  • 105.Davis KE, D. Neinast M, Sun K, et al. The sexually dimorphic role of adipose and adipocyte estrogen receptors in modulating adipose tissue expansion, inflammation, and fibrosis. Mol Metab. 2013;2(3):227–242. doi: 10.1016/j.molmet.2013.05.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Richard AJ, White U, Elks CM, Stephens JM. Adipose Tissue: Physiology to Metabolic Dysfunction. In: Feingold KR, Anawalt B, Blackman MR, et al. , eds. Endotext. MDText.com, Inc.; 2000. Accessed February 7, 2024. http://www.ncbi.nlm.nih.gov/books/NBK555602/ [Google Scholar]
  • 107.Trayhurn P, Beattie JH. Physiological role of adipose tissue: white adipose tissue as an endocrine and secretory organ. Proc Nutr Soc. 2001;60(3):329–339. doi: 10.1079/pns200194 [DOI] [PubMed] [Google Scholar]
  • 108.Cannon B, Nedergaard J. Brown adipose tissue: function and physiological significance. Physiol Rev. 2004;84(1):277–359. doi: 10.1152/physrev.00015.2003 [DOI] [PubMed] [Google Scholar]
  • 109.Carpentier AC, Blondin DP, Virtanen KA, Richard D, Haman F, Turcotte ÉE. Brown Adipose Tissue Energy Metabolism in Humans. Front Endocrinol. 2018;9:447. doi: 10.3389/fendo.2018.00447 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Heilbronn LK, de Jonge L, Frisard MI, et al. Effect of 6-mo. calorie restriction on biomarkers of longevity, metabolic adaptation and oxidative stress in overweight subjects. JAMA J Am Med Assoc. 2006;295(13):1539–1548. doi: 10.1001/jama.295.13.1539 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Neff LM, Hoffmann ME, Zeiss DM, et al. Core body temperature is lower in postmenopausal women than premenopausal women: potential implications for energy metabolism and midlife weight gain. Cardiovasc Endocrinol. 2016;5(4):151–154. doi: 10.1097/XCE.0000000000000078 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Torres Irizarry VC, Jiang Y, He Y, Xu P. Hypothalamic Estrogen Signaling and Adipose Tissue Metabolism in Energy Homeostasis. Front Endocrinol. 2022;13:898139. doi: 10.3389/fendo.2022.898139 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Santos RS, Frank AP, Fátima LA, Palmer BF, Öz OK, Clegg DJ. Activation of estrogen receptor alpha induces beiging of adipocytes. Mol Metab. 2018;18:51–59. doi: 10.1016/j.molmet.2018.09.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Koźniewski K, Wąsowski M, Jonas MI, et al. Epigenetic Regulation of Estrogen Receptor Genes’ Expressions in Adipose Tissue in the Course of Obesity. Int J Mol Sci. 2022;23(11):5989. doi: 10.3390/ijms23115989 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Lapid K, Lim A, Clegg DJ, Zeve D, Graff JM. Oestrogen signalling in white adipose progenitor cells inhibits differentiation into brown adipose and smooth muscle cells. Nat Commun. 2014;5:5196. doi: 10.1038/ncomms6196 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Schaffler MB, Kennedy OD. Osteocyte Signaling in Bone. Curr Osteoporos Rep. 2012;10(2):118–125. doi: 10.1007/s11914-012-0105-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Matsuoka K, Park K ae, Ito M, Ikeda K, Takeshita S. Osteoclast-Derived Complement Component 3a Stimulates Osteoblast Differentiation. J Bone Miner Res. 2014;29(7):1522–1530. doi: 10.1002/jbmr.2187 [DOI] [PubMed] [Google Scholar]
  • 118.Bonewald LF, Mundy GR. Role of Transforming Growth Factor Beta in Bone Remodeling: A Review. Connect Tissue Res. 1989;23(2–3):201–208. doi: 10.3109/03008208909002418 [DOI] [PubMed] [Google Scholar]
  • 119.Cauley JA. Estrogen and bone health in men and women. Steroids. 2015;99(Pt A):11–15. doi: 10.1016/j.steroids.2014.12.010 [DOI] [PubMed] [Google Scholar]
  • 120.Ji MX, Yu Q. Primary osteoporosis in postmenopausal women. Chronic Dis Transl Med. 2015;1(1):9–13. doi: 10.1016/j.cdtm.2015.02.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Cheng CH, Chen LR, Chen KH. Osteoporosis Due to Hormone Imbalance: An Overview of the Effects of Estrogen Deficiency and Glucocorticoid Overuse on Bone Turnover. Int J Mol Sci. 2022;23(3):1376. doi: 10.3390/ijms23031376 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Sioka C, Fotopoulos A, Georgiou A, Xourgia X, Papadopoulos A, Kalef-Ezra JA. Age at menarche, age at menopause and duration of fertility as risk factors for osteoporosis. Climacteric J Int Menopause Soc. 2010;13(1):63–71. doi: 10.3109/13697130903075337 [DOI] [PubMed] [Google Scholar]
  • 123.Couse JF, Lindzey J, Grandien K, Gustafsson JÅ, 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. doi: 10.1210/endo.138.11.5496 [DOI] [PubMed] [Google Scholar]
  • 124.Wiik A, Ekman M, Johansson O, Jansson E, Esbjörnsson M. Expression of both oestrogen receptor alpha and beta in human skeletal muscle tissue. Histochem Cell Biol. 2009;131(2):181–189. doi: 10.1007/s00418-008-0512-x [DOI] [PubMed] [Google Scholar]
  • 125.Hevener AL, Ribas V, Moore TM, Zhou Z. The Impact of Skeletal Muscle ERα on Mitochondrial Function and Metabolic Health. Endocrinology. 2020;161(2):bqz017. doi: 10.1210/endocr/bqz017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Ribas V, Drew BG, Zhou Z, et al. Skeletal muscle action of estrogen receptor α is critical for the maintenance of mitochondrial function and metabolic homeostasis in females. Sci Transl Med. 2016;8(334):334ra54–334ra54. doi: 10.1126/scitranslmed.aad3815 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Torres MJ, Kew KA, Ryan TE, et al. 17β-Estradiol Directly Lowers Mitochondrial Membrane Microviscosity and Improves Bioenergetic Function in Skeletal Muscle. Cell Metab. 2018;27(1):167–179.e7. doi: 10.1016/j.cmet.2017.10.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Hansen RD, Raja C, Baber RJ, Lieberman D, Allen BJ. Effects of 20-mg oestradiol implanttherapy on bone mineral density, fat distribution and musclemass in postmenopausal women. Acta Diabetol. 2003;40(1):s191–s195. doi: 10.1007/s00592-003-0063-5 [DOI] [PubMed] [Google Scholar]
  • 129.Ronkainen PHA, Kovanen V, Alén M, et al. Postmenopausal hormone replacement therapy modifies skeletal muscle composition and function: a study with monozygotic twin pairs. J Appl Physiol. 2009;107(1):25–33. doi: 10.1152/japplphysiol.91518.2008 [DOI] [PubMed] [Google Scholar]
  • 130.Erkkola R, Holma P, Järvi T, et al. Transdermal oestrogen replacement therapy in a Finnish population. Maturitas. 1991;13(4):275–281. doi: 10.1016/0378-5122(91)90236-j [DOI] [PubMed] [Google Scholar]
  • 131.Scharf MB, McDannold MD, Stover R, Zaretsky N, Berkowitz DV. Effects of estrogen replacement therapy on rates of cyclic alternating patterns and hot-flush events during sleep in postmenopausal women: a pilot study. Clin Ther. 1997;19(2):304–311. doi: 10.1016/S0149-2918(97)80118-X [DOI] [PubMed] [Google Scholar]
  • 132.Banks E, Beral V, Reeves G, Balkwill A, Barnes I, Million Women Study Collaborators. Fracture incidence in relation to the pattern of use of hormone therapy in postmenopausal women. JAMA. 2004;291(18):2212–2220. doi: 10.1001/jama.291.18.2212 [DOI] [PubMed] [Google Scholar]
  • 133.Cameron CR, Cohen S, Sewell K, Lee M. The Art of Hormone Replacement Therapy (HRT) in Menopause Management. J Pharm Pract. Published online April 1, 2023:8971900231167925. doi: 10.1177/08971900231167925 [DOI] [PubMed] [Google Scholar]
  • 134.Notelovitz M, Lenihan JP, McDermott M, Kerber IJ, Nanavati N, Arce J. Initial 17beta-estradiol dose for treating vasomotor symptoms. Obstet Gynecol. 2000;95(5):726–731. doi: 10.1016/s0029-7844(99)00643-2 [DOI] [PubMed] [Google Scholar]
  • 135.MacLennan A, Lester S, Moore V. Oral oestrogen replacement therapy versus placebo for hot flushes. Cochrane Database Syst Rev. 2001;(1):CD002978. doi: 10.1002/14651858.CD002978 [DOI] [PubMed] [Google Scholar]
  • 136.Goldstein I Recognizing and Treating Urogenital Atrophy in Postmenopausal Women. J Womens Health. 2010;19(3):425–432. doi: 10.1089/jwh.2009.1384 [DOI] [PubMed] [Google Scholar]
  • 137.de Novaes Soares C, Almeida OP, Joffe H, Cohen LS. Efficacy of Estradiol for the Treatment of Depressive Disorders in Perimenopausal Women: A Double-blind, Randomized, Placebo-Controlled Trial. Arch Gen Psychiatry. 2001;58(6):529–534. doi: 10.1001/archpsyc.58.6.529 [DOI] [PubMed] [Google Scholar]
  • 138.Costa GBC, Carneiro G, Umeda L, Pardini D, Zanella MT. Influence of Menopausal Hormone Therapy on Body Composition and Metabolic Parameters. BioResearch Open Access. 2020;9(1):80–85. doi: 10.1089/biores.2019.0050 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Cappelletti M, Wallen K. Increasing women’s sexual desire: The comparative effectiveness of estrogens and androgens. Horm Behav. 2016;78:178–193. doi: 10.1016/j.yhbeh.2015.11.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Cheng YS, Tseng PT, Wu MK, et al. Pharmacologic and hormonal treatments for menopausal sleep disturbances: A network meta-analysis of 43 randomized controlled trials and 32,271 menopausal women. Sleep Med Rev. 2021;57:101469. doi: 10.1016/j.smrv.2021.101469 [DOI] [PubMed] [Google Scholar]
  • 141.Pan Z, Wen S, Qiao X, Yang M, Shen X, Xu L. Different regimens of menopausal hormone therapy for improving sleep quality: a systematic review and meta-analysis. Menopause N Y N. 2022;29(5):627–635. doi: 10.1097/GME.0000000000001945 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Lufkin EG, Wahner HW, O’Fallon WM, et al. Treatment of postmenopausal osteoporosis with transdermal estrogen. Ann Intern Med. 1992;117(1):1–9. doi: 10.7326/0003-4819-117-1-1 [DOI] [PubMed] [Google Scholar]
  • 143.Lees B, Stevenson JC. The Prevention of Osteoporosis Using Sequential Low-Dose Hormone Replacement Therapy with Estradiol-17β and Dydrogesterone. Osteoporos Int. 2001;12(4):251–258. doi: 10.1007/s001980170113 [DOI] [PubMed] [Google Scholar]
  • 144.Cagnacci A, Venier M. The Controversial History of Hormone Replacement Therapy. Medicina (Mex). 2019;55(9):602. doi: 10.3390/medicina55090602 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Buist DSM, Newton KM, Miglioretti DL, et al. Hormone therapy prescribing patterns in the United States. Obstet Gynecol. 2004;104(5 Pt 1):1042–1050. doi: 10.1097/01.AOG.0000143826.38439.af [DOI] [PubMed] [Google Scholar]
  • 146.Clanget C, Hinke V, Lange S, Fricke R, Botko R, Pfeilschifter J. Patterns of hormone replacement therapy in a population-based cohort of postmenopausal German women. Changes after HERS II and WHI. Exp Clin Endocrinol Diabetes Off J Ger Soc Endocrinol Ger Diabetes Assoc. 2005;113(9):529–533. doi: 10.1055/s-2005-865802 [DOI] [PubMed] [Google Scholar]
  • 147.Guay MP, Dragomir A, Pilon D, Moride Y, Perreault S. Changes in pattern of use, clinical characteristics and persistence rate of hormone replacement therapy among postmenopausal women after the WHI publication. Pharmacoepidemiol Drug Saf. 2007;16(1):17–27. doi: 10.1002/pds.1273 [DOI] [PubMed] [Google Scholar]
  • 148.Menon U, Burnell M, Sharma A, et al. Decline in use of hormone therapy among postmenopausal women in the United Kingdom. Menopause N Y N. 2007;14(3 Pt 1):462–467. doi: 10.1097/01.gme.0000243569.70946.9d [DOI] [PubMed] [Google Scholar]
  • 149.Ettinger B, Wang SM, Leslie RS, et al. Evolution of postmenopausal hormone therapy between 2002 and 2009. Menopause. 2012;19(6):610. doi: 10.1097/gme.0b013e31823a3e5d [DOI] [PubMed] [Google Scholar]
  • 150.Mikkola TS, Tuomikoski P, Lyytinen H, et al. Increased Cardiovascular Mortality Risk in Women Discontinuing Postmenopausal Hormone Therapy. J Clin Endocrinol Metab. 2015;100(12):4588–4594. doi: 10.1210/jc.2015-1864 [DOI] [PubMed] [Google Scholar]
  • 151.Park CY, Lim JY, Kim WH, Kim SY, Park HY. Evaluation of menopausal hormone therapy use in Korea (2002–2013): A nationwide cohort study. Maturitas. 2021;146:57–62. doi: 10.1016/j.maturitas.2021.02.003 [DOI] [PubMed] [Google Scholar]
  • 152.Clarkson TB, Meléndez GC, Appt SE. Timing hypothesis for postmenopausal hormone therapy: its origin, current status, and future. Menopause N Y N. 2013;20(3):342–353. doi: 10.1097/GME.0b013e3182843aad [DOI] [PubMed] [Google Scholar]
  • 153.El Khoudary SR, Aggarwal B, Beckie TM, et al. Menopause Transition and Cardiovascular Disease Risk: Implications for Timing of Early Prevention: A Scientific Statement From the American Heart Association. Circulation. 2020;142(25):e506–e532. doi: 10.1161/CIR.0000000000000912 [DOI] [PubMed] [Google Scholar]
  • 154.Hodis HN, Mack WJ, Henderson VW, et al. Vascular Effects of Early versus Late Postmenopausal Treatment with Estradiol. N Engl J Med. 2016;374(13):1221–1231. doi: 10.1056/NEJMoa1505241 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Ali N, Sohail R, Jaffer SR, et al. The Role of Estrogen Therapy as a Protective Factor for Alzheimer’s Disease and Dementia in Postmenopausal Women: A Comprehensive Review of the Literature. Cureus. 2023;15(8):e43053. doi: 10.7759/cureus.43053 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Chlebowski RT, Hendrix SL, Langer RD, et al. Influence of Estrogen Plus Progestin on Breast Cancer and Mammography in Healthy Postmenopausal WomenThe Women’s Health Initiative Randomized Trial. JAMA. 2003;289(24):3243–3253. doi: 10.1001/jama.289.24.3243 [DOI] [PubMed] [Google Scholar]
  • 157.Gold EB, Colvin A, Avis N, et al. Longitudinal Analysis of the Association Between Vasomotor Symptoms and Race/Ethnicity Across the Menopausal Transition: Study of Women’s Health Across the Nation. Am J Public Health. 2006;96(7):1226–1235. doi: 10.2105/AJPH.2005.066936 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Appling S, Paez K, Allen J. Ethnicity and vasomotor symptoms in postmenopausal women. J Womens Health 2002. 2007;16(8):1130–1138. doi: 10.1089/jwh.2006.0033 [DOI] [PubMed] [Google Scholar]
  • 159.DeBono NL, Robinson WR, Lund JL, et al. Race, Menopausal Hormone Therapy, and Invasive Breast Cancer in the Carolina Breast Cancer Study. J Womens Health. 2018;27(3):377–386. doi: 10.1089/jwh.2016.6063 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Cui J, Shen Y, Li R. Estrogen synthesis and signaling pathways during ageing: from periphery to brain. Trends Mol Med. 2013;19(3):197–209. doi: 10.1016/j.molmed.2012.12.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Hou N, Hong S, Wang W, Olopade OI, Dignam JJ, Huo D. Hormone Replacement Therapy and Breast Cancer: Heterogeneous Risks by Race, Weight, and Breast Density. JNCI J Natl Cancer Inst. 2013;105(18):1365–1372. doi: 10.1093/jnci/djt207 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Chlebowski RT, Barrington W, Aragaki AK, et al. Estrogen Alone and Health Outcomes in Black Women by African Ancestry: A Secondary Analyses of a Randomized Controlled Trial. Menopause N Y N. 2017;24(2):133–141. doi: 10.1097/GME.0000000000000733 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Graham S, Archer DF, Simon JA, Ohleth KM, Bernick B. Review of menopausal hormone therapy with estradiol and progesterone versus other estrogens and progestins. Gynecol Endocrinol. 2022;38(11):891–910. doi: 10.1080/09513590.2022.2118254 [DOI] [PubMed] [Google Scholar]
  • 164.Gompel A, Stuenkel CA. Neurokinin 3 receptor antagonists for menopausal vasomotor symptoms, an appraisal. Cell Rep Med. 2023;4(6):101076. doi: 10.1016/j.xcrm.2023.101076 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Navarro VM, Gottsch ML, Chavkin C, Okamura H, Clifton DK, Steiner RA. Regulation of Gonadotropin-Releasing Hormone Secretion by Kisspeptin/Dynorphin/Neurokinin B Neurons in the Arcuate Nucleus of the Mouse. J Neurosci. 2009;29(38):11859–11866. doi: 10.1523/JNEUROSCI.1569-09.2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Clarkson J, Han SY, Piet R, et al. Definition of the hypothalamic GnRH pulse generator in mice. Proc Natl Acad Sci U S A. 2017;114(47):E10216–E10223. doi: 10.1073/pnas.1713897114 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Rance NE, Dacks PA, Mittelman-Smith MA, Romanovsky AA, Krajewski-Hall SJ. Modulation of body temperature and LH secretion by hypothalamic KNDy (kisspeptin, neurokinin B and dynorphin) neurons: A novel hypothesis on the mechanism of hot flushes. Front Neuroendocrinol. 2013;34(3):10.1016/j.yfrne.2013.07.003. doi: 10.1016/j.yfrne.2013.07.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.David PS, Smith TL, Nordhues HC, Kling JM. A Clinical Review on Paroxetine and Emerging Therapies for the Treatment of Vasomotor Symptoms. Int J Womens Health. 2022;14:353–361. doi: 10.2147/IJWH.S282396 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Chen MN, Lin CC, Liu CF. Efficacy of phytoestrogens for menopausal symptoms: a meta-analysis and systematic review. Climacteric. 2015;18(2):260–269. doi: 10.3109/13697137.2014.966241 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Patisaul HB, Jefferson W. The pros and cons of phytoestrogens. Front Neuroendocrinol. 2010;31(4):400–419. doi: 10.1016/j.yfrne.2010.03.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Martinkovich S, Shah D, Planey SL, Arnott JA. Selective estrogen receptor modulators: tissue specificity and clinical utility. Clin Interv Aging. 2014;9:1437–1452. doi: 10.2147/CIA.S66690 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Júnior JK, Kulak CAM, Taylor HS. SERMs in the prevention and treatment of postmenopausal osteoporosis: an update. Arq Bras Endocrinol Metabol. 2010;54(2):200–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Emons G, Mustea A, Tempfer C. Tamoxifen and Endometrial Cancer: A Janus-Headed Drug. Cancers. 2020;12(9):2535. doi: 10.3390/cancers12092535 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Mortimer JE, Flatt SW, Parker BA, et al. Tamoxifen, hot flashes and recurrence in breast cancer. Breast Cancer Res Treat. 2008;108(3):421–426. doi: 10.1007/s10549-007-9612-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Balfour JA, Goa KL. Raloxifene. Drugs Aging. 1998;12(4):335–341. doi: 10.2165/00002512-199812040-00006 [DOI] [PubMed] [Google Scholar]
  • 176.Mosca L, Grady D, Barrett-Connor E, et al. Effect of Raloxifene on Stroke and Venous Thromboembolism According to Subgroups in Postmenopausal Women at Increased Risk of Coronary Heart Disease. Stroke. 2009;40(1):147–155. doi: 10.1161/STROKEAHA.108.518621 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Palacios S, Farias MLF, Luebbert H, et al. Raloxifene is not associated with biologically relevant changes in hot flushes in postmenopausal women for whom therapy is appropriate. Am J Obstet Gynecol. 2004;191(1):121–131. doi: 10.1016/j.ajog.2003.10.701 [DOI] [PubMed] [Google Scholar]
  • 178.Lello S, Capozzi A, Scambia G. The Tissue-Selective Estrogen Complex (Bazedoxifene/Conjugated Estrogens) for the Treatment of Menopause. Int J Endocrinol. 2017;2017:5064725. doi: 10.1155/2017/5064725 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Pinkerton JV, Pickar JH, Racketa J, Mirkin S. Bazedoxifene/conjugated estrogens for menopausal symptom treatment and osteoporosis prevention. Climacteric. 2012;15(5):411–418. doi: 10.3109/13697137.2012.696289 [DOI] [PubMed] [Google Scholar]
  • 180.Shin JJ, Kim SK, Lee JR, Suh CS. Ospemifene: A Novel Option for the Treatment of Vulvovaginal Atrophy. J Menopausal Med. 2017;23(2):79–84. doi: 10.6118/jmm.2017.23.2.79 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Goldstein SR, Scheele WH, Rajagopalan SK, Wilkie JL, Walsh BW, Parsons AK. A 12-month comparative study of raloxifene, estrogen, and placebo on the postmenopausal endometrium11Eli Lilly and Company sponsored this study. Jorge Londoño, MD, Aarti Shah, PhD, and Anura Abeyratne, PhD, contributed to the study design and analysis. Mayme Wong, PhD, Leo Plouffe, Jr, MD, Sheryl L. Silfen, MD, and Peter Kenemans, MD, contributed to the interpretation of the results, made content suggestions, and critically reviewed the manuscript. Obstet Gynecol. 2000;95(1):95–103. doi: 10.1016/S0029-7844(99)00502-5 [DOI] [PubMed] [Google Scholar]
  • 182.Prokai L, Nguyen V, Szarka S, et al. The prodrug DHED selectively delivers 17β-estradiol to the brain for treating estrogen-responsive disorders. Sci Transl Med. 2015;7(297):297ra113. doi: 10.1126/scitranslmed.aab1290 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Salinero AE, Abi-Ghanem C, Venkataganesh H, et al. Brain Specific Estrogen Ameliorates Cognitive Effects of Surgical Menopause in Mice. bioRxiv. Published online August 13, 2023:2023.08.09.552687. doi: 10.1101/2023.08.09.552687 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Madak-Erdogan Z, Kim SH, Gong P, et al. Design of pathway-preferential estrogens that provide beneficial metabolic and vascular effects without stimulating reproductive tissues. Sci Signal. 2016;9(429):ra53. doi: 10.1126/scisignal.aad8170 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Zuo Q, Chen KL, Arredondo Eve A, et al. Pathway Preferential Estrogens Prevent Hepatosteatosis Due to Ovariectomy and High-Fat Diets. Nutrients. 2021;13(10):3334. doi: 10.3390/nu13103334 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Gérard C, Foidart JM. Estetrol: From Preclinical to Clinical Pharmacology and Advances in the Understanding of the Molecular Mechanism of Action. Drugs RD. 2023;23(2):77–92. doi: 10.1007/s40268-023-00419-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Holinka CF, Brincat M, Coelingh Bennink HJT. Preventive effect of oral estetrol in a menopausal hot flush model. Climacteric J Int Menopause Soc. 2008;11 Suppl 1:15–21. doi: 10.1080/13697130701822807 [DOI] [PubMed] [Google Scholar]
  • 188.Gaspard U, Taziaux M, Mawet M, et al. A multicenter, randomized study to select the minimum effective dose of estetrol (E4) in postmenopausal women (E4Relief): part 1. Vasomotor symptoms and overall safety. Menopause N Y N. 2020;27(8):848–857. doi: 10.1097/GME.0000000000001561 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Takahashi K, Manabe A, Okada M, Kurioka H, Kanasaki H, Miyazaki K. Efficacy and safety of oral estriol for managing postmenopausal symptoms. Maturitas. 2000;34(2):169–177. doi: 10.1016/S0378-5122(99)00108-5 [DOI] [PubMed] [Google Scholar]
  • 190.Griesser H, Skonietzki S, Fischer T, Fielder K, Suesskind M. Low dose estriol pessaries for the treatment of vaginal atrophy: A double-blind placebo-controlled trial investigating the efficacy of pessaries containing 0.2mg and 0.03mg estriol. Maturitas. 2012;71(4):360–368. doi: 10.1016/j.maturitas.2011.12.022 [DOI] [PubMed] [Google Scholar]
  • 191.Sánchez-Rovira P, Hirschberg AL, Gil-Gil M, Bermejo-De Las Heras B, Nieto-Magro C. A Phase II Prospective, Randomized, Double-Blind, Placebo-Controlled and Multicenter Clinical Trial to Assess the Safety of 0.005% Estriol Vaginal Gel in Hormone Receptor-Positive Postmenopausal Women with Early Stage Breast Cancer in Treatment with Aromatase Inhibitor in the Adjuvant Setting. The Oncologist. 2020;25(12):e1846–1854. doi: 10.1634/theoncologist.2020-0417 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Abdi F, Mobedi H, Mosaffa N, Dolatian M, Ramezani Tehrani F. Hormone Therapy for Relieving Postmenopausal Vasomotor Symptoms: A Systematic Review. Arch Iran Med. 2016;19:141–146. [PubMed] [Google Scholar]
  • 193.Lovre D, Lindsey SH, Mauvais-Jarvis F. Effect of menopausal hormone therapy on components of the metabolic syndrome. Ther Adv Cardiovasc Dis. 2016;11(1):33–43. doi: 10.1177/1753944716649358 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Raz L, Hunter L, Dowling NM, et al. Differential Effects of Hormone Therapy on Serotonin, Vascular Function and Mood in the KEEPS. Climacteric J Int Menopause Soc. 2016;19(1):49–59. doi: 10.3109/13697137.2015.1116504 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Fait T Menopause hormone therapy: latest developments and clinical practice. Drugs Context. 2019;8:212551. doi: 10.7573/dic.212551 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Gosset A, Pouillès JM, Trémollieres F. Menopausal hormone therapy for the management of osteoporosis. Best Pract Res Clin Endocrinol Metab. 2021;35(6):101551. doi: 10.1016/j.beem.2021.101551 [DOI] [PubMed] [Google Scholar]
  • 197.Meziou N, Scholfield C, Taylor CA, Armstrong HL. Hormone therapy for sexual function in perimenopausal and postmenopausal women: a systematic review and meta-analysis update. Menopause N Y N. 2023;30(6):659–671. doi: 10.1097/GME.0000000000002185 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Wright AC, Beaudoin FL, McQueen RB, et al. The effectiveness and value of fezolinetant for moderate-to-severe vasomotor symptoms associated with menopause: A summary from the Institute for Clinical and Economic Review’s Midwest Public Advisory Council. J Manag Care Spec Pharm. 2023;29(6):10.18553/jmcp.2023.29.6.692. doi: 10.18553/jmcp.2023.29.6.692 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Yang D, Li J, Yuan Z, Liu X. Effect of Hormone Replacement Therapy on Cardiovascular Outcomes: A Meta-Analysis of Randomized Controlled Trials. PLoS ONE. 2013;8(5):e62329. doi: 10.1371/journal.pone.0062329 [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES