Abstract
The hypothalamic-pituitary-ovarian (HPO) axis is a complex endocrine feedback mechanism controlling ovulation in female vertebrates. Balance of the HPO axis requires correct secretion of sex steroids from the ovarian follicle to inhibit release of gonadotropins from the pituitary. Several conditions of ovarian dysfunction such as menopause, primary ovarian insufficiency, and polycystic ovary syndrome involve imbalances in the HPO axis, contributing to infertility. Intriguingly, these disorders also share a higher incidence of cognitive and emotional dysregulations, as well as a heightened risk of certain neurodegenerative conditions with age. It is understood that estradiol exerts neuroprotective functions, but gonadotropin signaling is less understood. High concentrations of circulating follicle-stimulating hormone (FSH) and luteinizing hormone (LH) have shown to contribute to neurodegenerative disease states, but are not addressed as part of traditional hormone replacement therapy. To identify the mechanistic connections between ovarian disorders and heightened susceptibility of the brain to pathological aging, a multisystem experimental approach is required, considering each HPO axis player as an individual effector. In this review, we will summarize current knowledge on the effects of estradiol, progesterone, FSH, and LH on neuronal susceptibility to pathology. We will describe ways in which the HPO axis becomes imbalanced during ovarian dysfunction, and how systemic inflammation can become an additional HPO axis effector. Finally, we will recommend solutions to the presented gaps in knowledge, and suggest avenues of future research to pursue development of therapeutics targeting both ovarian and brain health in patients.
Keywords: sex hormones, gonadotropins, neuroprotection, neurodegeneration, ovarian disorders, endocrine dysfunction
Ovarian disorders describe conditions affecting the health of an ovary, often affecting fertility and quality of life. Common symptoms include infertility, vaginal discharge, pelvic discomfort, and weight gain, as well as a host of neurological symptoms including hot flashes, insomnia, mood swings, and brain fog (1-4). Ovarian health can decline due to germline mutations to the oocyte, somatic mutations to oocyte maturation machinery (5), environmental stressors, inflammation, and oxidative stress, among others (6). These conditions are exacerbated by a disruption of the balance of the hypothalamic-pituitary-ovarian (HPO) axis, an endocrine feedback loop in which gonadotropins from the pituitary stimulate sex steroid release in the ovarian follicle. Poor follicular quality often affects the ability of the follicle to respond to gonadotropin cues, causing an imbalance in the HPO axis that contributes both to fertility outcomes and overall symptomatology (7).
A less understood aspect of ovarian disorders is their frequent co-occurrence with neurological conditions, particularly a higher prevalence of dementias later in life, which this review will explore (8). For the purposes of this review, based on terminologies of cited studies, we will refer to people with ovaries as “women”; however, we acknowledge transgender men and nonbinary people also experience these issues. Despite the large proportion of women living with ovarian disorders, the causes of these neurological symptoms remain elusive and understudied. HPO axis imbalance is implicated as a driving factor for neurological phenotypes. A considerable body of literature describes estrogens as neuroprotective, thus their loss would suggest an increased susceptibility to neuronal disorders (9-11). However, direct supplementation of estrogen to patients post menopause has failed to decrease risk of dementias past age 65 years (12). Additionally, elevated serum gonadotropins such as luteinizing hormone (LH) are implicated in the promotion of dementia phenotypes, demonstrating the complexity of the HPO axis in modulating brain health (13). This review focuses on the current body of knowledge surrounding ovarian disorders with regard to the HPO axis, and roles of the HPO axis in susceptibility to dementias. We will then speculate on possible mechanistic connections between ovarian disorder characteristics and resulting neuronal phenotypes, and propose future work in the field to elucidate these mechanisms.
Ovarian Disorders
Hypothalamic-Pituitary-Ovarian Axis
Reproductive maturity is established in women on onset of menarche, when the HPO axis becomes active. Growth cues to the brain stimulate a population of neurons in the hypothalamus to secrete gonadotropin-releasing hormone (GnRH) (14). GnRH triggers pituitary release of gonadotropins, namely follicle-stimulating hormone (FSH), to act on ovarian follicles and trigger their growth (15). FSH is required for proper oocyte maturation, largely due to its action on granulosa cells (GCs), the cells that directly surround the oocyte within the follicle. GCs are the primary producers of estradiol, the dominant estrogen during reproductive years. It is a sex steroid responsible for promoting oocyte growth and proliferation of the GCs (14, 16, 17). Successful follicle growth eventually leads to ovulation, an event triggered by pituitary release of another gonadotropin, LH. The ovulated egg travels to the fallopian tube to await fertilization, leaving the GCs to luteinize and form the corpus luteum, a postovulatory structure (18). LH triggers the corpus luteum to secrete progesterone, a sex steroid responsible for maintaining the health of the egg post ovulation. Through endocrine action, estradiol and progesterone both bind to GnRH regulator neurons, as well as gonadotropic pituitary cells to inhibit release of FSH and LH. This negative feedback prevents further estradiol and progesterone production by the GCs, resetting the menstrual cycle (19).
Androgens are the precursor sex steroids to estradiol. Testosterone, an androgen, is the primary sex steroid in males, and is converted to estradiol in the ovary by aromatase. Progesterone is one of the precursors of testosterone production, greatly contributing to androgen synthesis. Although understudied, androgens are implicated in neuroprotective roles, but can also contribute to ovarian disorders when imbalanced (20, 21).
Not in the purview of this review, Insulin-like growth factor (IGF), the somatotropic axis, (14) and the transforming growth factor-β superfamily play roles in fine-tuning the timing and stages of ovulation (22). IGF will be discussed later in the context of insulin resistance contributions to pathology.
Hypothalamic-Pituitary-Ovarian Axis Disorders in the Ovary
Ovarian disorders are triggered by a wide variety of mechanisms. To achieve proper follicular function, the oocyte must develop correctly in utero, accomplish accurate chromosome segregation, then in the adult mammal respond to growth cues, and secrete prosurvival factors to encourage GC proliferation (23). Steroidogenesis is initiated by gonadotropins and growth factors, and requires sensitivity to gonadotropins to produce sex steroids (14). Secreted steroids must then exert function on brain structures to modulate further release on gonadotropins, or ovulation will not occur (19).
Outlined in Table 1, mutations in genes essential to these processes are linked to several types of ovarian disorders (26, 29, 32, 35). Inflammation can also greatly affect GC-oocyte communication. High oxidative stress conditions in the ovaries of patients with polycystic ovary syndrome (PCOS) can prevent efficient energy production in the GCs, inducing apoptosis and reducing oocyte quality (36, 37). Resulting inflammation from mitochondrial damage and other factors was found to correlate inversely both with steroid biosynthesis rates and embryo quality (38).
Table 1.
Summary of epidemiology and symptomatology of ovarian disorders
Condition | Prevalence in US | Age of onset, y | Symptomatology | Comorbidities/Complications | HPO axis balance | Genetic linkage examples |
---|---|---|---|---|---|---|
Menopause | 1.3 million more women per y in US, eventually 100% of women will experience menopause (24). | Mean of 52 in US (25). | Irregular periods, vaginal dryness, hot flashes, night sweats, mood changes, brain fog, insomnia, fatigue (4) | Increased risk of cardiovascular disease, stroke, diabetes, osteoporosis, dementia (4) | High FSH, high LH Low estradiol, low progesterone, low testosterone, low AMH (4). |
PALB2, ETAA1, HROB, BRCA, CHEK2 (26) |
POI | 3.5% globally as of 2021 (27) | Most commonly ages 35-40, globally (1) | Irregular periods before age 40, hot flashes, night sweats, vaginal dryness, mood changes, fatigue, joint pain, loss of focus or memory (1) | Increased risk of dementia, Parkinson disease, heart disease, osteoporosis, hypothyroidism (28). | High FSH, high LH, low estradiol, low progesterone, low AMH, low testosterone (1) | FMR1, PGRMC1, AR, FOXO4, BMP15, FS, HR, STAR, CYP17A1, BMPR1B, GJA4, INHA, GDF9, ESR1 (29). |
PCOS | 4%-10% of reproductive-aged women worldwide (3) | Majority of instances ages 20-30 y, globally (3) | Menstrual irregularities, heavy periods, hirsutism, acne, insulin sensitivity, obesity, oily skin, fatigue, mood changes, headaches, insomnia (3) | Increased risk of heart disease, endometrial hyperplasia, uterine cancer, diabetes, dementia (3) | High androgens, high LH, high free testosterone, high AMH. If ovulations are not consistent, low progesterone and high estradiol (30, 31) | CYP family, AR, AMH, FSHR, LH, INS, IRS-1, IRS2 (32), THADA (33), DENND1A (34) |
Abbreviations: AMH, antimüllerian hormone; FSH, follicle-stimulating hormone; HPO, hypothalamic-pituitary-ovarian; LH, luteinizing hormone; PCOS, polycystic ovary syndrome; POI, primary ovarian insufficiency.
This is not an exhaustive list of conditions that could be considered ovarian disorders. We have omitted infertility conditions arising from other organs such as hypopituitarism and hyperthyroidism/hypothyroidism (5). The effects of ovarian cancer on the HPO axis are difficult to parse due to noted effects of chemotherapy on cognition not necessarily arising from the cancer itself, so we have omitted these from our list (39).
Endometriosis is an intriguing example of ovarian dysfunction in which the uterine lining forms ectopic lesions on any pelvic organ, including the ovaries. These lesions produce estradiol de novo, increasing circulating estradiol levels. Ovarian endometriosis often induces accelerated follicular atresia due to inflammation, raising FSH levels (2, 40). A Taiwanese study found that patients with endometriosis were 2 times more likely than control individuals to experience a mental disorder, including anxiety, depression, bipolar, sleep disorders, psychotic disorders, and dementia (41). While we can speculate on a potential role for endocrine dyscrasia in the case of brain alterations in ovarian endometriosis, it is difficult to parse this apart from the more established roles of mental illness and chronic pain in increasing susceptibility of the brain to dementias (42). Surprisingly, asymptomatic endometriosis patients were also found to have structural alterations in the brain (43). More research will be required to parse the role of hormonal imbalance in the symptomatology of ovarian endometriosis.
Menopause
Menopause occurs when loss of oocyte quality from aging accelerates follicular atresia, leaving insufficient follicle counts to produce enough sex steroids for ovulation (24). Healthy women in the United States experience menopause at age 52 years on average (25). The precise timing of menopause is highly hereditary, with several gene mutations linked to onset of symptoms. Intriguingly, most of these genes exhibit roles in DNA damage repair responses, with beneficial variants also increasing risks of certain cancers (26).
Early menopause is defined as a loss of ovarian function occurring between ages 40 and 45 years. Onset of symptoms begins in perimenopause, the transition period into menopause. During this transition, FSH rises, exacerbating follicle maturation (4). Estradiol and progesterone levels become more variable as the follicle pool dwindles, contributing to symptoms. Changes to the brain begin during perimenopause, including decreases in white matter and glucose metabolism in patients (44). Menopause is medically confirmed 12 months after the last period (4). Symptoms lessen during postmenopause, with brain changes stabilizing over time (44).
Menopause increases the risk for comorbidities through several mechanisms. Core body temperature of women post menopause is 0.25 °C cooler on average, causing a 3.25% drop in energy expenditure that would result in 6.7 pounds of weight gain per year in the average woman (45). Increased central adiposity promotes inflammation, contributing to insulin resistance in some patients (46). Menopause increases the risk of dementias and other cognitive symptoms with age, with hormone replacement therapy (HRT) ameliorating this risk only before age 65 years (12). Because of these and other risk factors, age of menopause was found to correlate with mortality and lifespan, with a 2.9% reduction in mortality for every year of later menopause (47, 48).
Primary Ovarian Insufficiency
Primary ovarian insufficiency (POI) is the absence or early depletion of the ovarian reserve, occurring before age 40 years (1). High FSH, low estradiol, and low antimüllerian hormone (AMH) are all primary characteristics of POI. AMH is produced by GCs and directly correlates with follicle abundance, making it a standard diagnostic tool for POI (49). POI affects around 3.5% of reproductive-aged women globally (27). Genetic factors contribute to around 10% to 15% of cases. X chromosome abnormalities, HPO axis receptors, and steroid producers are among identified mutations (29).
Symptoms arising from POI are extremely similar to those experienced by women in perimenopause. Comorbidities are also similar, including greater risk cardiovascular disease and dementias (28, 29). One study found low-grade chronic inflammation associated with POI, exacerbating aging phenotypes (50). Multiple studies have determined a correlation between age at menopause and lower dementia risk (8). One study in China found that women with a younger age of natural menopause were significantly more likely to experience mild cognitive impairment, as well as receive a dementia diagnosis later in life (51). Patients are often prescribed HRT both of estrogen and progesterone to combat these risks and symptoms (29).
Polycystic Ovary Syndrome
PCOS is the most common endocrinopathy affecting premenopausal women, with around a 4% to 10% prevalence worldwide (3). PCOS is characterized by hyperandrogenism and high LH (30, 31). Other diagnostic criteria include amenorrhea and inflammatory, fluid-filled cysts present on the ovary. Theca cell steroidogenesis becomes intrinsically activated, aberrantly producing androgens, which modulate FSH signaling in GCs to trigger proliferation. Higher AMH and high follicle counts are reported in PCOS. Impaired timing of ovulation from sex steroid imbalance leads to fluid buildup in follicular structures, creating cysts. Chronic inflammation further propagates androgen production in the follicle (3). Although PCOS is a multifactorial disease, familial studies suggest inheritance may occur in an X-linked polygenic manner (32, 52). Mutations associated with PCOS development include gonadotropin genes, AMH, and insulin resistance genes (32).
One study found that by midlife, women with PCOS diagnosis performed worse on fluency tests, and had less white matter integrity than age-matched controls (6). A Canadian study found prevalence of dementia in patients with PCOS to be 2 times higher than in controls, with the median age of diagnosis 19 years younger in patients with PCOS (53). Dietary therapies and oral contraceptives are most commonly used to treat symptoms of PCOS, aiming to reduce free androgen concentrations and improve insulin resistance (32).
Brain Aging and Neurodegeneration
Hallmarks of Disease
Neurodegenerative diseases are disorders in which a progressive loss of neurons occurs in the central or peripheral nervous system. As neurons are terminally differentiated cells, maintenance of their health is carefully controlled by numerous mechanisms (54). Disruptions to homeostatic systems are considered hallmarks of neurodegeneration, and were described thoroughly in Wilson et al in 2023 (54). We will briefly describe hallmarks relevant to our topic, including proteostasis and energy metabolism.
Intracellular neuronal health is controlled by autophagy and proteostasis networks. Without recycling of disordered proteins and damaged organelles, aggregates stress intracellular machinery and cytoskeletal transport. Cytoskeletal modulation is particularly important in neurons due to axon length, making efficient transport of nutrients between the synapse and dendrites paramount for neuronal survival and signaling. Aggregates can be deposited in the extracellular space or even passed to other neurons through prion-like spread in extracellular vesicles, causing stress to whole subregions of neurons (54). In Alzheimer disease (AD), protein aggregates of amyloid beta (Aβ) plaques and tau tangles are hallmarks of disease (55).
Control of synaptic signaling is essential for functionality of neuronal networks. Energy supplied by mitochondria allows neurotransmitter release and reuptake. Mitochondrial dysfunction leads to an excess of intracellular calcium, triggering neurotransmitter release to the synaptic cleft that cannot be reabsorbed by the neuron without calcium efflux. This causes an overactivation of neurotransmitter receptors on the receiving neuron. This overactivation, especially by excitatory neurotransmitters like glutamate, can prompt excess calcium influx into neighboring healthy neurons. This phenotype is called “excitotoxicity” and results in the release of reactive oxygen species (ROS), energy depletion, and neuronal death. Proper maintenance of mitochondrial health and neurotransmitter resorption is essential to prevent this feedback (54).
Neuroinflammation
The brain comprises neurons, glial cells, neuronal stem cells, and blood vessels. While neurons are responsible for information processing, glia perform vital support roles to maintain the health of brain tissue. Microglia are a macrophage-lineage brain-resident immune cell, exerting housekeeping and defensive functions. Overactivation of microglia drives neuroinflammation in several neurodegenerative disorders, causing chronic neuronal stress and eventual cell death. Astrocytes accomplish numerous neuroprotective tasks, including damage responses, neurotransmitter clearance from the synaptic cleft, and maintenance of the blood-brain barrier (BBB). Astrocytes become overactivated in response to excessive stimuli, altering their ability to modulate neurotransmitter recycling and promoting excitotoxicity (54).
The BBB is a vascular barrier composed of endothelial cells, tight junctions, gap junctions, pericytes, and astrocytes. Endothelial cells use specialized transporters to allow the passage of nutrients and oxygen into the brain, while preventing entry of circulating ROS, toxins, and cytokines. Smaller, highly lipophilic molecules such as estradiol can freely cross the BBB, whereas larger, hydrophilic molecules like FSH and LH may require an active transport mechanism to cross (56). Endothelial cells are the most sensitive to damage from inflammatory molecules. Chronic, low-grade inflammation worsens the integrity of endothelial tight junctions over time (57). Astrocytes project endfeet to the endothelial cells, and release numerous factors including IGF1 and apolipoprotein-E to regulate distribution of occludins, claudins, and connexins. Reactive astrocytes downregulate expression of tight junction proteins of the BBB, promoting leakage and exacerbating neuroinflammation. BBB leakage also arises from damage to endothelial cells or neuroinflammation, causing infiltration of immune cells (58) and toxins into the brain, contributing to further neuroinflammation and predisposing the brain to cognitive disorders (54).
Limbic System
Neurodegeneration can involve disruptions in any or all of the described systems, but some brain regions, such as the limbic system, are especially susceptible to initial damages. Comprising the diencephalon and lower cortex, the limbic system is a highly interconnected region responsible for memory consolidation, emotional and social processing, motivation, and learning (59).
An especially vulnerable limbic structure is the hippocampus, the primary site of adult neurogenesis in the brain. The hippocampus is responsible for learning and memory tasks, specifically memory recall. During healthy aging, the hippocampus experiences one of the steepest declines in volume compared to other brain regions, rivaled only by cerebral white matter (60). The hippocampus is also especially sensitive to hypoxia, making strokes and cardiovascular disease an especially important risk factor for dementia development (61). The hippocampus contains a subpopulation of pyramidal neurons required for learning that have especially high energy demands and an abundance of glucocorticoid receptors, making them extremely sensitive to mitochondrial dysfunction, excitotoxicity, oxygen deficiency, ROS, and toxins (62).
The hypothalamus, also within the limbic system, controls bodily homeostasis through prompting hormone release, controlling sleep-wake cycles, body temperature, and satiety. It is also highly susceptible to damage, with one study finding gene expression signatures with age to be the most changed in the hypothalamus of mice (63). Another study found astrocytes in the hypothalamus were more transcriptionally altered than cortical neurons, potentially hindering their immune-modulating processes (64). In presymptomatic patients with AD, Huntington disease, and amyotrophic lateral sclerosis, hypothalamic alterations related to energy metabolism and caloric intake were altered, suggesting the hypothalamus to be especially vulnerable to early pathologies (65).
The prefrontal cortex is responsible for behavioral regulation, attention, focus, and executive functions. Most publications consider at least the ventromedial frontal cortical regions to be part of the limbic system (66). This area is specifically vulnerable to mitochondrial dysfunction with age. In ovariectomized (OVX) monkeys, morphological changes to mitochondria, as well as increased ROS and oxidative stress, were present in the prefrontal cortex. This effect was rescued by estradiol treatment (67), suggesting a loss of estradiol due to HPO axis dysfunction could affect normal operations. Along with increased susceptibility to disease, the hippocampus, hypothalamus, and limbic system as a whole share high expression of gonadotropin receptors (68), aromatase, and estrogen receptors (69), hinting at possible perturbations to signaling after drastic hormonal change.
Endocrine Action on Brain Homeostasis
Sex Steroids
Sex steroids include estrogens, progesterone, and testosterone. Their neurobiological actions are detailed in Table 2. We will focus specifically on the estrogen estradiol, the dominant form of estrogen during reproductive years in both sexes (88, 89). Sex steroids in the brain either enter from circulation or are produced locally by neurons. Neuronally derived estradiol (NDE) has unique effects from circulating estradiol, responding directly to injury, promoting cell survival through brain-derived neurotrophic factor (BDNF) expression, an important neuroprotective factor, and preventing neuroinflammation (70). Aromatase expression is required for NDE production, and is most highly expressed in the limbic system, suggesting damage to the limbic system could lower NDE (69) and promote disease.
Table 2.
Neuroprotective actions of sex steroids in literature
Sex steroid effector | Neuroprotective activity |
---|---|
NDE | BDNF induction, learning promotion, prevention of gliosis following injury: As identified in mouse models lacking aromatase activity in the brain, NDE is important for increasing forebrain synaptic density, efficient synaptic transmission and long-term potentiation, cognitive function, and promoting BDNF expression. NDE also protects against impairment following ischemic brain injury, promotes astrocyte repair mechanisms, and prevents reactive microgliosis. NDE release by neurons and astrocytes is increased following brain injury, likely as a protective mechanism (70). |
Estradiol | BDNF induction: Estradiol binds an estrogen response element on the BDNF gene, and induces phosphorylation of CREB, both activating and amplifying BDNF to promote neuronal survival, neurogenesis, synapse formation, and synaptic plasticity, especially evident in the hippocampus (10). |
Estradiol | Cell survival, antioxidant activity: In a study observing the effects of ethanol intoxication on the brain of rats, estradiol was found to activate Sirtuin1 to lower NFκB and TNF-α, preventing neuroinflammation. Estradiol also increased expression of Nrf2 and Ho1 to protect against oxidative stress. Estradiol itself can also scavenge free radicals. Estradiol inhibits neuronal apoptosis by inhibiting caspase 3 and Bax (9), and promoting antiapoptotic effectors B-cell lymphoma (Bcl)-xL and Bcl-w (71). Estrogen receptors are found in neuronal mitochondria, suggesting direct action of estradiol on mitochondria to activate NRF1 and protect against oxidative stress (11). |
Estradiol | Stroke protection: To prevent onset of ischemic stroke, estradiol can widen blood vessels. Stroke can mechanically damage neurons surrounding the blood vessel, an event that increases risk of dementia onset by 2 to 5 times (72, 73). |
Estradiol | Prevents protein aggregation: In the context of AD development, estradiol increases PP2a, causing dephosphorylation of tau and stabilizing the cytoskeleton. Estradiol inhibits BACE1, the enzyme that cleaves APP to its pathological form, the antecedent to amyloid plaque formation (69). Estradiol promotes expression of neprilysin, an enzyme that degrades amyloid (74). |
Estradiol | BBB integrity protection: Estradiol can directly act on cerebral vasculature to suppress inflammatory markers and reduce immune cell adhesion (75). Estradiol promotes survival of endothelial cells, protecting them against inflammation and hypoxia-induced apoptosis (11). Estradiol also acts through ANXA1 to promote BBB integrity. ANXA1 stabilizes cytoskeletal connections in the BBB, prevents circulating immune cell entry, and reduces neuroinflammation. Estradiol mediation of ANXA1 promotes expression of occludin and zona-occludens-1, and reduces expression of ICAM-1 and VCAM-1 to prevent leukocyte invasion (76). |
Estradiol | Prevent insulin resistance: Estradiol acts through IGF-1 to promote glucose transport and uptake in the brain, with IGF-1 decreasing as estradiol is lost with age, and neuroprotective actions of estradiol hindered without presence of IGF-1 (11). |
Estradiol | Prevents neuroinflammation: Estradiol acts through ANXA1 to promote a proresolving microglial phenotype, enhancing phagocytosis and anti-inflammatory signatures such as the mannose receptor, CD-206 (77). |
Progesterone | Cell survival: Progesterone activates MAPK/ERK and PI3K/Akt pathways to promote neuronal survival, and activates BDNF (78). |
Progesterone | Prevent neuroinflammation: In a multiple sclerosis murine model, progesterone inhibited microgliosis and prevented release of proinflammatory cytokines (79). In a murine model of autoimmune encephalomyelitis, progesterone treatment reduced proinflammatory mediators CD-11b, TNF-α, and lowered activity of nitric oxide synthase, a precursor to oxidative stress (80). Progesterone downregulates inflammasome formation and TNF expression in microglia and induces BDNF release and autophagy in astrocytes (81). |
Progesterone | Promotes BBB integrity: In a rat ischemia model, progesterone was found to increase tight junction expression through occludin 1 and claudin 5, and reduced inflammation by downregulating metalloproteinases (82). |
Testosterone | Prevents cell death under stress: Testosterone partially rescued cell death in neuronal cultures experiencing hypoxia and glucose deprivation, dependent on binding to the androgen receptor (83). |
Testosterone | Increases synaptic density through BDNF induction: Testosterone acts through the androgen receptor to rescue dendritic atrophy in spinal motor neurons after loss of neighboring neuron connections (84). In mice, testosterone can enhance spatial performance, likely due to androgen receptors present in hippocampal pyramidal neurons (85). Testosterone rescued synaptic density in the hippocampus of gonadectomized male mice by inducing expression of BDNF (86). |
Testosterone | Prevent protein aggregation: Testosterone can downregulate BACE1 to prevent amyloid plaque deposition (87) |
Abbreviations: AD, Alzheimer disease; ANXA1, annexin A1; APP, amyloid precursor protein; BACE1, β-secretase; Bax, Bcl-2 associated X-protein; BBB, blood-brain barrier; BDNF, brain-derived neurotrophic factor; cAMP, cyclic adenosine monophosphate; CD, cluster of differentiation; CREB, cAMP response element binding protein; Ho1, heme oxygenase-1; ICAM-1, intercellular adhesion molecule 1; IGF-1, insulin-like growth factor 1; MAPK/ERK, mitogen-activated protein kinase/extracellular signal-regulated kinase; NDE, neuronally derived estradiol; NFκB, nuclear factor-κB; NRF1, nuclear respiratory factor-1; Nrf2, nuclear factor erythroid 2-related factor 2; PI3K/Akt, phosphoinositide 3-kinases/protein kinase B; PP2a, protein phosphatase 2A; TNF-α, tumor necrosis factor-α; VCAM-1, vascular cell adhesion molecule-1.
Research on the neuroprotective effects of estradiol rarely separate effects between NDE and circulating estradiol; however, one can assume that findings from the following research can largely be attributed to the higher concentration of circulating estradiol sourced from the ovaries. Estradiol promotes BDNF signaling induction (10), acts as an antioxidant and promotes cell survival (9, 11), protects against stroke damage (72, 73), prevents insulin resistance (11), and prevents protein aggregation of AD hallmarks Aβ (71) and phosphorylated tau (pTau) tangles (69, 74), effectively protecting neurons from energy loss and excitotoxicity. Intriguingly, female patients with AD have significantly lower levels of estradiol than age-matched controls (90).
Progesterone is also neuroprotective, promoting cell survival through similar mechanisms to estradiol, such as promoting cell survival and reducing neuroinflammation (78). However, progesterone-estradiol combination therapy can cause both positive and negative effects on brain pathology, suggesting the molecules can act both synergistically and antagonistically. In a study testing the effect of estradiol and progesterone on mitochondrial output, estradiol and progesterone could separately rescue mitochondrial function after poisoning, but this effect was lost with coadministration (91). Progesterone can inhibit estrogen receptor expression, as well as prevent estrogen receptors from binding to chromatin to exert their effects. It is unclear in which scenarios this may or may not be occurring, or if another mechanism of action is responsible for this antagonism (78).
Testosterone exhibits some neuroprotective qualities as well, preventing cell death (83), protein aggregation (87), and increasing synaptic density (84-86). However, free testosterone concentration was found to correlate with impaired cognition in women with PCOS, specifically in psychomotor speed and visuospatial learning, suggesting an essential balance of sex steroids to accomplish efficient cognitive function (92).
Regulating neuroinflammation is an essential protective mechanism in the brain. Estradiol and progesterone can both reduce neuroinflammation (77, 79-81 ) and strengthen junctions of the BBB (11, 75, 76, 82). Excessive doses of testosterone in male rats reduced expression of claudin-5 and occludin (93); however, chronic depletion of testosterone also reduced expression of claudin-5, zona occludens-1 (ZO1), and upregulated inflammatory factors tumor necrosis factor (TNF), inducible nitric oxide synthase, and cyclooxygenase-2, suggesting a balance of testosterone concentration is required to maintain BBB integrity (94).
Gonadotropins
Gonadotropins refer to FSH and LH, which are cyclically released from the pituitary. Their neurobiological actions are detailed in Table 3. Both can become chronically high with loss of sex steroid feedback (15).
Table 3.
Neurobiological effects of gonadotropins in literature
Gonadotropin effector | Neurobiological activity |
---|---|
FSH | Increased depressive behavior and neuroinflammation: Researchers found impaired memory, increased anxiety and loss of pleasure seeking during behavioral testing of male mice administered FSH, and increased TNF-α, IL-6, and activated glia in mouse brains compared to controls. Knockdown of FSHR in the hippocampus reversed these effects (95). In AD mice, treatment with FSH increased GFAP and IBA1, markers for activated astrocytes and microglia, respectively. In OVX mice, these signatures increased as well, with this effect rescued by treatment with an FSH-inhibiting antibody. This effect was also rescued by estradiol treatment (96). |
FSH | Synapse loss: Synaptic markers synapsin and synaptophysin were decreased following FSH treatment in male mice, with glutamatergic synapse markers GluR1 and VGluT1 decreased as well. Knockdown of FSHR in the hippocampus reversed these effects (95). |
FSH | Increased pathological aggregates: Experiments in AD mice revealed FSH acts independently of other HPO axis players to bind FSHR in the hippocampus and activate CCAAT/enhancer binding protein β–asparaginyl endopeptidase (C/EBPβ–AEP) to initiate δ-secretase activity, increasing cleavage of APP to its pathological forms. FSH dosing also induced tau phosphorylation and pTau accumulation, as well as amyloid plaque accumulation, and loss of dendritic spines (68). |
LH | Correlation with cognitive deficiency: In males, LH concentration was correlated with poor memory recall, a mechanism of the hippocampus (97). In male and female patients with AD, LH concentrations were higher than age-matched controls, with LH correlating with cognitive decline, amyloid plaque load, and tau tangles (13). |
LH | Increased pathological aggregates: Treating neuroblastoma cells with LH increased their secretion of Aβ, due to an increased activity of BACE1, cleaving APP to its pathological form. As Aβ plaque formation can drive hyperphosphorylation of tau, this would further exacerbate AD pathologies (98). Experiments in OVX mice, guinea pigs, and human embryonic stem cells saw increases in APP when treated with LH (99). |
LH | Worsened cognition: Transgenic mice overexpressing LH had worse cognitive performance on a maze task compared to wild-types; this effect was reversed when LH receptors were nonfunctional (81). When OVX mice treated with estradiol were also treated with an LH homologue, their performance on cognitive and behavioral tests significantly worsened (100). |
LH | Promotion of steroidogenesis: When treated with LH, rat hippocampal cells and human neuroblastoma cells saw increased pregnenolone productions through upregulation of steroidogenic acute regulatory protein (101). As pregnenolone is a sex steroid precursor and itself neuroprotective, this suggests a nuanced role for LH in disease susceptibility. |
FSH + LH | Reducing BDNF and impairing cognition: Gonadotropin inhibition via leuprolide acetate in AD mice improved their cognition in behavioral tasks and reduced their Aβ plaque load (102). Similarly, treatment with a GnRH antagonist in OVX rats increased BDNF expression, rescuing cognition (103). |
FSH + LH | BBB modulation: OVX in mice greatly increased permeability of the BBB, coupled by dysregulation of Cx43, a gap junction. Researchers reported that while OVX increased intensity of Cx43 staining, gonadotropin inhibition lowered Cx43 expression. However, it is unclear what the physiological consequence of this change would be, as Cx43 is vital for gap and tight junction organization, and requires both adequate expression and correct localization to exert proper functionality (104). |
Abbreviations: Aβ, amyloid β; AD, Alzheimer disease; APP, amyloid precursor protein; BBB, blood-brain barrier; BDNF, brain-derived neurotrophic factor; Cx43, connexin-43; FSH, follicle-stimulating hormone; FSHR, follicle-stimulating hormone receptor; GFAP, glial fibrillary acidic protein; GluR1, glutamate receptor 1; GnRH, gonadotropin-releasing hormone; HPO, hypothalamic-pituitary-ovarian; IBA1, ionized calcium-binding adaptor molecule 1; IL, interleukin; LH, luteinizing hormone; OVX, ovariectomized; TNF-α, tumor necrosis factor-α; VGluT1, vesicular glutamate transporter 1.
LH exists in two populations—one is released from the pituitary and circulates peripherally, while the other is produced from the hypothalamus and regulates expression levels of LH receptor (LHR) in the brain (105). Peripheral LH levels correlate with AD progression, Aβ load, and cognitive decline in humans (106, 107), but it is unclear if this effect is through a direct action of LH on neurons after crossing the BBB, an indirect action by which high peripheral concentrations of LH suppress brain synthesis of LH, or an entrance of peripheral LH to the brain only after damage to the BBB. Multiple earlier works have found LH to cross the BBB to a limited extent; it is unknown if this action would change under a disease state or state of endocrine dyscrasia (108, 109).
It is clear that high peripheral LH is associated with negative neurological outcomes. In postmenopausal women, high LH correlated with poor cognitive performance (110). Older women with AD had higher levels both of LH and FSH compared to age-matched controls (106). In animal studies, LH administration induces cognitive deficits and increases amyloid deposition (100, 111). Intriguingly, studies directly testing the effect of LH on neuronal cells find mixed results. In neuronal culture, LH increased production of plaques via increased β-secretase (BACE1) activity, but also increased production of pregnenolone, a precursor to the neuroprotective estradiol (98, 101). When administered directly to the mouse brain, one study found LH to impair memory function (112). A separate study found this administration to ameliorate cognitive OVX symptoms (113). Notably, the former study used a much higher dose than the latter. Considering LH can dose-dependently downregulate expression of its receptor, this suggests that benefit or harm could depend greatly on dose. It has been shown that peripheral and brain LH have inverse concentrations naturally (114). Whether or not LH crosses the BBB, high peripheral LH would therefore yield low signaling through LHR, either through suppression of brain LH or through suppression of LHR expression (115). LHR is highly expressed in many disease-sensitive brain regions, including the hippocampus, hypothalamus, cortex, pons, cerebellum, thalamus, and medulla (116), and is an important contributor to memory processes and long-term potentiation through extracellular signal-regulated kinase (ERK) and protein kinase A signaling (117). A reduction in LHR signaling due to high peripheral LH is a plausible theory behind human studies correlating high LH with neurodegenerative indicators. A third possibility is that peripheral LH weakens the BBB, allowing for its own entry to the brain. In OVX mice, FSH and LH inhibition rescued a leaky BBB, indicating one or both of these molecules could allow for its own entry to the brain (104). This would suggest that other factors like toxins or inflammatory cytokines could increase brain concentrations of gonadotropins. Future studies should explore this and other possibilities. For further reading on the subject, see the 2021 review by Mey et al (105).
Many studies correlate FSH concentrations with negative consequences on the brain, but a suggested mechanism has only recently been proposed in the literature. In postmenopausal women, FSH levels correlated with vascular stiffness, increasing stroke risk (118), as well as concentrations of inflammatory cytokines (119). In premenopausal women, FSH levels correlated with depression risk, which itself is a risk factor for dementias (120, 121). FSH receptor (FSHR) brain expression is described in multiple recent studies, with transcripts identified in the mouse cerebellum, olfactory bulb, hippocampus, cerebral cortex, medulla, midbrain and pons, forebrain, thalamus, and hypothalamus (68, 122). Another study validated findings in the yak hypothalamus, pineal gland (123), and mouse hippocampus (124). A further study found FSHR expression in the human cortex, and the mouse cortex and hippocampus (122). Similarly to LH, it is unclear if FSH is able to cross the BBB, if it exerts actions indirectly, or if it contributes to BBB destruction to enter the brain. A recent study using an AD mouse model used a fluorescent tag to observe injected FSH crossing the BBB. The researchers then showed that deleterious effects of FSH were occurring through FSHR binding and subsequent activation of the CCATT/enhancer binding proteins beta and delta (C/EBPβ/δ)-secretase pathway, including deposition of amyloid plaques and tau tangles. They also showed increased gliosis signatures, suggesting FSH modulation of microglia (68). The physical properties of FSH suggest it would have to be actively transported across the BBB, and more research will be necessary to parse how and through what circumstances endothelial transport of FSH would occur. Several studies report FSH can induce inflammatory states in immune cells in peripheral tissues (125, 126), suggesting damage to the BBB through inflammation could be another mode of entry. Overall, more research in the coming years will be required to highlight either a similar or disparate mechanism of FSH action on the brain compared to LH, with current but limited studies hinting at a deleterious and direct action of FSH on sensitive brain regions. For further reading on the topic, see Xue et al (127).
Although neurotypical women are at higher risk of developing dementia than men, men with Down syndrome are more likely to develop the disease than women with Down syndrome. In both cases, the higher risk group experiences elevated concentrations of FSH and LH, suggesting a direct link between gonadotropin concentration and disease risk, regardless of sex (102). Gonadotropin inhibition can induce BDNF expression and rescue cognition (102, 103). More research is required to parse potential regulation of FSH and LH on each other in the brain, with regard to modulation of receptor expression in disease-sensitive brain regions.
Ovarian Disorders and Brain Pathology
This review has described common symptoms and comorbidities of ovarian disorders, and key brain regions susceptible to disease. It is remarkable that many of these symptoms share dysregulation of limbic system structures, a stress-susceptible area heavily populated by gonadotropin receptors (128, 129). Common factors dysregulating activity of these brain structures would heavily implicate HPO axis involvement in symptomatology across the disorders. This is summarized in Fig. 1.
Figure 1.
Common symptoms of ovarian disorders share involvement of limbic systems. A, Brain regions involved or altered in depression (130). B, Brain regions involved or altered in mood disorders (131). C, Brain regions involved or altered during hot flashes (132-135). D, Brain regions involved in sleep and altered in insomniac patients (136-139). E, Brain regions involved or altered in conditions using the broad term “brain fog” to describe cognitive symptoms (140-143). Abbreviations: AC, anterior cerebellum; ACC, anterior cingulate cortex; Amyg, amygdala; CC, cingulate cortex; CN, caudate nucleus; CV, cerebellar vermis; DLPFC, dorsolateral prefrontal cortex; GP, globus pallidus; HC, hippocampus; Hypo, hypothalamus; INS, insular cortex; MCC, medial cingulate cortex; MFG, middle frontal gyrus; MPFC, medial prefrontal cortex; OFC, orbitofrontal cortex; PAG, periaqueductal gray; PCC, posterior cingulate cortex; PFC, prefrontal cortex; RC, rhinal cortex; SN, substantia nigra; Str, striatum; Thal, thalamus.
Specific studies highlighting the risk factors discussed in the following sections are displayed in Table 4.
Table 4.
Neurological risk factors for disease development arising from ovarian conditions, with evidence from literature
Ovarian condition | Risk factor for brain pathology | Studies |
---|---|---|
Menopause/POI | Lower endogenous estradiol | A study on women with Down syndrome found a correlation between lower endogenous levels of estradiol post-menopause and higher risk of developing AD (144). Estradiol modulates serotonin signaling (145), and low levels of estradiol-target BDNF are correlated with depression onset (146). |
Menopause/POI | High LH and FSH blood concentrations | One study in men found that circulating LH concentration directly correlated with circulating levels of Aβ(1-42) (147). A study in women ages 40 to 65 years found blood FSH and LH levels to be positively correlated to Aβ load and negatively correlated with gray matter volume in the frontal cortex (148). In multiple studies, FSH level positively correlated with onset of depression. It is possible that FSH receptors on monocytes signal to produce proinflammatory cytokines, affecting mood (149). Although less is known about LH in the context of mood modulation, one study on rats found that a gonadotropin inhibitor reduced anxious behaviors following ovariectomy (150). |
Menopause/POI/PCOS | Insulin resistance. | Evidence of insulin dysregulation was found in hippocampal and cortical sections of patients with AD (151). IGF-1 can promote vasodilation to protect against stroke, promote integrity of the BBB, promote anti-inflammatory glial states, and enhance insulin signaling and glucose uptake by cells (152). Loss of estradiol and IGF-1 and increases in cortisol and insulin resistance predisposing women to stroke onset (55). Insulin resistance also promotes storage of fat in nonadipose tissues and increases appetite, predisposing patients to obesity (153, 154). Insulin resistance promotes vasoconstriction and stroke onset. Insulin itself promotes clearance of amyloid plaques and tau tangles (153, 154). Hyperinsulinemia can cause adipose tissues to become inflamed and release inflammatory cytokines, with this effect amplified in individuals with obesity (155). |
Menopause/POI | High cortisol. | Cortisol binds glucocorticoid receptors in the hippocampus and other areas. Chronic activation of the glucocorticoid receptors was found to damage hippocampal neurons by activation of NLRP1 (156). Levels of cortisol in patients with AD correlated with disease severity and progression (157). Cortisol administration to AD mice increased BACE1, Aβ deposition, and tau hyperphosphorylation (158). |
Menopause/POI | Chronic inflammation | In patients with POI, serum concentrations of IL-6, IL-8, IL-17, and TNF-α increase with age in patients (159). Similarly, women undergoing menopause had significantly higher serum levels of IL-1β, IL-8, and TNF-α than in fertile women (160). |
Menopause | Structural changes. | During the menopause transition, women were found to experience cerebral decreases in white matter, glucose metabolism, and increases in cerebral blood flow and Aβ load. Metabolism shifted to fatty acid utilization, possibly exacerbated by growing insulin resistance. Cognition was not affected during this transition, suggesting compensatory mechanisms occurring as a response to HPO axis dysregulation (44). Mouse models of menopause using OVX mice found a decrease in acetylcholine neuron density, an increase in anxiety, Aβ load, and decrease in locomotion (161). |
PCOS | Hyperandrogenism | Androgen treatment to young biological females was found to increase spatial cognition, but worsen verbal fluency (162). Another study correlated high levels of free testosterone in patients with PCOS to worse verbal fluency, verbal memory, and visuospatial memory than controls (92). Several other studies report mixed results, with one finding testosterone correlated with better performance on visuospatial tasks and frontal gyrus connectivity networks, while others report worse performance on spatial visualization, and no effect on cognition. Overall, women with PCOS were found to perform worse on tasks of verbal fluency, verbal learning, verbal memory, visuospatial and executive functions, spatial reasoning, auditory processing, and finger dexterity than controls (163). Treating rats with a nonaromatizable androgen elevated serum levels of TNF-α and IL-1β compared to controls (164). In adipocytes, androgens affect phosphorylation of PKC, downregulating glucose transport and conferring an effect of insulin resistance on metabolism. In turn, hyperinsulinemia stimulates the ovaries to produce more androgens, propagating the cycle (165). |
Abbreviations: Aβ, amyloid β; AD, Alzheimer disease; APP, amyloid precursor protein; BBB, blood-brain barrier; BDNF, brain-derived neurotrophic factor; FSH, follicle-stimulating hormone; FSHR, follicle-stimulating hormone receptor; HPO, hypothalamic-pituitary-ovarian; IGF-1, insulin-like growth factor 1; IL, interleukin; LH, luteinizing hormone; NLRP1, NLR family pyrin domain containing 1; OVX, ovariectomized; PCOS, polycystic ovary syndrome; PKC, protein kinase C; POI, primary ovarian insufficiency; TNF-α, tumor necrosis factor-α.
Menopause and Primary Ovarian Insufficiency
Menopause and POI, or premature menopause, are characterized by high concentrations of serum gonadotropins and low concentrations of sex steroids. Insulin resistance, loss of IGF-1, and heightened cortisol often occur during this transition as well (4). Estradiol and progesterone prevent neuroinflammation and induce neuronal survival and energy homeostasis (144-146), with their loss likely promoting pathology (9-11, 76, 78, 81).
However, HRT post menopause has historically yielded conflicting results in reducing AD risk, with one study concluding that the greatest benefits are yielded if treatment begins in perimenopause, and continues to age 65 years (12). This may suggest that dysregulation of other HPO axis players is also a significant contributor to disease risk. FSH and LH rise considerably during early perimenopause, and may promote protein aggregation (148) and neuroinflammation in animal models (68, 95, 96, 98, 99). Although mechanisms are still being parsed, numerous correlational and human studies and mechanistic animal studies conclude high levels of peripheral LH and FSH, like what occurs during menopause, are detrimental to the health of sensitive brain regions (68, 100, 104, 106, 110, 111, 119).
Additionally, IGF-1 loss, insulin resistance, stroke risk, and cortisol levels all contribute to disease susceptibility. Insulin resistance prevents effective transport of glucose into cells, promoting senescence and hyperglycemia. This causes impaired neuronal glucose metabolism (151), weakened synaptic plasticity, excitotoxicity of glutaminergic neurons, and improper vasodilation of blood vessels (55). Insulin resistance is primarily driven by declines in estradiol, poor sleep and stress, although not all women experience insulin resistance post menopause. Slowing of metabolism leads to increased central adiposity and increased inflammation, contributing to BBB leakage (45, 46). Cortisol also rises in some women during menopause, and can lead to pathological protein aggregation (156-158). IGF-1 loss with age is also attributed to estradiol loss, increasing pathologies of insulin resistance (55, 152).
POI is correlated with oxidative stress and inflammatory aging (159), especially in the case of autoimmune POI, in which the immune system attacks the ovaries. Similarly, women undergoing premature menopause had significantly higher serum cytokines than fertile women (160). This can damage the BBB and promote neuroinflammation.
Besides dementia risk (161), other cognitive effects, such as depression and mood changes, are seen as early as the time of POI diagnosis. The limbic system, which controls emotional responses, is especially sensitive to neuroinflammation and damage (166). High FSH and LH can negatively affect mood (149, 150). Limbic system stress, neuroinflammation, and estradiol loss also contribute to brain fog, insomnia, and hot flashes (167). A combination of high gonadotropins and loss of BBB integrity could potentially exacerbate neuronal symptoms.
Polycystic Ovary Syndrome
PCOS is characterized most commonly by high androgens, high LH, and high free testosterone. Sometimes patients also experience low progesterone if ovulations are infrequent, and high estrogen due to increased aromatization of androgens. It is speculated that hyperandrogenic conditions desensitize negative feedback mechanisms of sex steroids and hyperexcite GnRH neurons. AMH also activates GnRH firing, perpetuating anovulation while the oocyte population remains high (168). High circulating LH can contribute to protein aggregation and cognitive decline (100, 106, 110, 111), with high estradiol potentially offering a counteracting effect (99, 100, 111).
The effects of hyperandrogenism on cognition are complex, with studies finding both benefits and hindrances on cognition (92, 162). One review summarizes that women with PCOS were found to perform worse on tasks of verbal fluency, verbal learning, verbal memory, visuospatial and executive functions, spatial reasoning, auditory processing, and finger dexterity than controls (163). Some of these effects could be due to hyperandrogenism; however, high LH and psychological stressors could also be contributors.
It is important to note that women with PCOS are more likely to experience depression, anxiety, bipolar disorder, and obsessive-compulsive disorder (169). Physical symptoms like weight gain, chronic pain, and infertility are major contributors to these diagnoses. Intriguingly, increased levels of free testosterone correlated with higher perceived stress in healthy women, suggesting hyperandrogenism could contribute to mental illness (170). Additionally, type 2 diabetic patients were found to have increased cortisol levels compared to healthy controls, suggesting an interplay between insulin resistance and stress (171). Psychiatric disorder diagnosis is associated with an increased risk of dementia development, with disruptions of neurotransmitter networks contributing to excitotoxicity and neuroinflammation (42). Future work should parse the contribution of endocrine imbalance to psychiatric disorder development, as an alternative contributor to dementia onset.
Insulin resistance is a common symptom of PCOS, affecting 65% to 95% of patients (165). Twelve weeks of androgen exposure in rats induced a state of insulin resistance, suggesting hyperandrogenism can induce this symptom (172). Insulin resistance impairs synaptic integrity, promotes stroke onset, and mitochondrial dysfunction of neurons (153, 154). Poor sleep in women with PCOS is directly correlated to insulin resistance, contributing to symptoms of brain fog and insomnia (167).
PCOS is a condition of chronic, low-grade inflammation (155, 164). Increased adipose tissue, insulin dysfunction, and glucose intake all promote further release of cytokines from circulating immune cells (173). This inflammation can weaken the BBB and may induce neuroinflammation.
Conclusions and Future Directions
This review highlights the distinct roles of gonadotropins and sex steroids in the brain and reproductive organs, urging a reconsideration of the HPO axis as a network of discrete hormonal effectors with multisystemic effect. Current therapies overlook the effects of ovarian dysfunction on brain health. We suggest an integrative, temporally sensitive, and mechanistically precise approach to understanding and intervening in reproductive aging and its neurological sequelae (Fig. 2).
Figure 2.
Summary of potential effects of ovarian disorders on brain pathology. A, Menopause and POI comprise inflammatory aging symptoms, with potential consequences of neuroinflammation through BBB disruption, gliosis, and loss of neuroprotection through sex steroid depletion, and pathological aggregation through high gonadotropin concentration. B, PCOS comprises metabolic dyscrasia and hyperandrogenism, with potential consequences of BBB disruption from systemic inflammation, altered cerebral energy metabolism through insulin resistance, and protein aggregation through high peripheral LH.Abbreviations: AMH, antimüllerian hormone; BBB, blood-brain barrier; BDNF, brain-derived neurotrophic factor; FSH, follicle-stimulating hormone; IGF-1, insulin-like growth factor 1; LH, luteinizing hormone; PCOS, polycystic ovary syndrome; POI, primary ovarian insufficiency.
HRT remains a cornerstone of menopausal care, but evidence suggests neuroprotective benefits diminish after age 65 years (12), demonstrating intervention should begin concurrently with initial hormone dyscrasia, requiring more careful monitoring of endocrine effectors at the suspected time of perimenopause. Additionally, estradiol and progesterone may execute counterproductive actions in the brain when administered together (78, 91). Progesterone administration aids in uterine cancer prevention (174); thus, it could be beneficial to administer hormones more cyclically, minimizing overlap between estrogen and progesterone concurrence. Mechanisms underlying the combined therapy on other organs must be better understood.
To optimize treatment strategies across the spectrum of ovarian dysfunction, future clinical trials should consider levels both of sex steroids and gonadotropins as therapeutic targets. Rebalancing the endocrine environment may be necessary to relieve the range of symptoms experienced in ovarian disorders. For example, gonadotropins rise markedly in perimenopause, making FSH and LH valuable markers for identifying critical windows for intervention. These trials should evaluate cognitive as well as ovarian health outcomes.
A critical next step in HPO axis research is clarifying the mechanistic action of gonadotropins in the aging brain, as questions of BBB access and direct modulation of neurons are inconsistent (113). Future work could prioritize questions of endothelial entry of gonadotropins, as well as the development of specific inhibitors for each gonadotropin, to clarify previous findings using leuprolide acetate to inhibit both gonadotropins simultaneously (102-104). Additionally, existing drugs approved for menopause care that inhibit GnRH pulsatility, such as fezolinetant (brand name VEOZAH, manufactured by Astellas Pharma US Inc.), could potentially be repurposed for improving dementia risk (175, 176). Future trials are needed to assess efficacy of these compounds in improving neuronal outcomes.
Concurrently, BBB disruption in ovarian dysfunction and the potential role of gonadotropins warrant closer study. While animal models of menopause have advanced our understanding of how chronic inflammation compromises BBB integrity (104), comparable models for POI and PCOS remain underdeveloped. Characterizing these models will allow better understanding for ways in which ovarian disorders can contribute to BBB disruption, as well as identifying targetable mechanisms for reinforcing BBB integrity. Therapeutic strategies that modulate local inflammation may also offer long-term benefits, such as using metformin, a diabetes drug, for improving insulin-resistance–derived inflammation in menopause and PCOS (177).
Finally, lifestyle interventions remain a low-risk and potentially high-reward adjunct to pharmacologic approaches. Recent studies have found that increasing dietary intake of fruits, fiber, unsaturated fats, vegetables, and lactobacillus-based probiotics increases beneficial populations of gut microbes, lowering inflammation and improving mood (178-182). Increasing physical activity also lowers vascular markers of inflammation (183) and can improve insulin resistance (184). As part of an integrative therapeutic model, such interventions should be tested in combination with targeted therapies to optimize health-span in individuals with ovarian hormone dysregulation.
In sum, advancing ovarian research requires a reconceptualization of the HPO axis as a network of individual yet interdependent effectors. A multisystem, syncretic experimental approach will be critical to move beyond symptom control and toward a future of personalized endocrine aging management.
Abbreviations
- Aβ
amyloid β
- AD
Alzheimer disease
- AMH
antimüllerian hormone
- ANXA1
annexin A1
- APP
amyloid precursor protein
- BACE1
β-secretase
- Bax
Bcl-2 associated X-protein
- BBB
blood-brain barrier
- Bcl
B-cell lymphoma
- BDNF
brain-derived neurotrophic factor
- cAMP
cyclic adenosine monophosphate
- C/EBPβ/δ
CCAAT/enhancer binding proteins beta and delta
- Cx43
connexin-43
- FSH
follicle-stimulating hormone
- FSHR
follicle-stimulating hormone receptor
- GC
granulosa cell
- GFAP
glial fibrillary acidic protein
- GluR1
glutamate receptor 1
- GnRH
gonadotropin-releasing hormone
- Ho1
heme oxygenase-1
- HPO
hypothalamic-pituitary-ovarian
- HRT
hormone replacement therapy
- IBA1
ionized calcium-binding adaptor molecule 1
- ICAM-1
intercellular adhesion molecule 1
- IGF-1
insulin-like growth factor 1
- IL
interleukin
- LH
luteinizing hormone
- LHR
luteinizing hormone receptor
- MAPK/ERK
mitogen-activated protein kinase/extracellular signal-regulated kinase
- NDE
neuronally derived estradiol
- NFκB
nuclear factor-κB
- NLRP1
NLR family pyrin domain containing 1
- NRF1
nuclear respiratory factor-1
- Nrf2
nuclear factor erythroid 2-related factor 2
- OVX
ovariectomized
- PCOS
polycystic ovary syndrome
- PI3K/AKT
phosphoinositide 3-kinases/protein kinase B
- PKC
protein kinase C
- POI
primary ovarian insufficiency
- PP2a
protein phosphatase 2A
- pTau
phosphorylated tau
- ROS
reactive oxygen species
- TNF-α
tumor necrosis factor-α
- VCAM1
vascular cell adhesion molecule-1
- VGlut1
vesicular glutamate transporter 1
- ZO1
zona occludens-1
Contributor Information
Heather Valera, Molecular Biology, Cell Biology, and Biochemistry Graduate Program, Brown University, Providence, RI 02906, USA; Department of Obstetrics and Gynecology, Program in Women's Oncology, Women and Infants Hospital of Rhode Island, Providence, RI 02905, USA.
Angela Chen, Department of Obstetrics and Gynecology, Program in Women's Oncology, Women and Infants Hospital of Rhode Island, Providence, RI 02905, USA; Division of Biology and Medicine, Brown University, Providence, RI 02906, USA.
Kathryn J Grive, Department of Obstetrics and Gynecology, Program in Women's Oncology, Women and Infants Hospital of Rhode Island, Providence, RI 02905, USA; Department of Obstetrics and Gynecology, Warren Alpert Medical School of Brown University, Providence, RI 02905, USA.
Funding
This work was supported by the National Institutes of Health, National Institute for Child Health and Development (No. R01-HD111322 to K.J.G.).
Disclosures
The authors declare that there are no conflicts of interest.
Data Availability
Data sharing is not applicable to this article as no data sets were generated or analyzed during the current study.
References
- 1. Federici S, Rossetti R, Moleri S, et al. Primary ovarian insufficiency: update on clinical and genetic findings. Front Endocrinol (Lausanne). 2024;15:1464803. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Chauhan S, More A, Chauhan V, Kathane A. Endometriosis: a review of clinical diagnosis, treatment, and pathogenesis. Cureus. 2022;14(9):e28864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Singh S, Pal N, Shubham S, et al. Polycystic ovary syndrome: etiology, current management, and future therapeutics. J Clin Med. 2023;12(4):1454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Davis SR, Pinkerton J, Santoro N, Simoncini T. Menopause-biology, consequences, supportive care, and therapeutic options. Cell. 2023;186(19):4038‐4058. [DOI] [PubMed] [Google Scholar]
- 5. Mikhael S, Punjala-Patel A, Gavrilova-Jordan L. Hypothalamic-pituitary-ovarian axis disorders impacting female fertility. Biomedicines. 2019;7(1):5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Huddleston HG, Jaswa EG, Casaletto KB, et al. Associations of polycystic ovary syndrome with indicators of brain health at midlife in the CARDIA cohort. Neurology. 2024;102(4):e208104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Cai J, Lou H-Y, Dong M-Y, et al. Poor ovarian response to gonadotropin stimulation is associated with low expression of follicle-stimulating hormone receptor in granulosa cells. Fertil Steril. 2007;87(6):1350‐1356. [DOI] [PubMed] [Google Scholar]
- 8. Pszczołowska M, Walczak K, Miśków W, et al. Association between female reproductive factors and risk of dementia. J Clin Med. 2024;13(10):2983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Khan M, Shah SA, Kim MO. 17β-Estradiol via SIRT1/Acetyl-p53/NF-kB signaling pathway rescued postnatal rat brain against acute ethanol intoxication. Mol Neurobiol. 2018;55(4):3067‐3078. [DOI] [PubMed] [Google Scholar]
- 10. Scharfman HE, MacLusky NJ. Estrogen and brain-derived neurotrophic factor (BDNF) in hippocampus: complexity of steroid hormone-growth factor interactions in the adult CNS. Front Neuroendocrinol. 2006;27(4):415‐435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Zárate S, Stevnsner T, Gredilla R. Role of estrogen and other sex hormones in brain aging. Neuroprotection and DNA repair. Front Aging Neurosci. 2017;9:430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Mosconi L, Nerattini M, Williams S, Fink M. New horizons in menopause, menopausal hormone therapy, and Alzheimer's disease: current insights and future directions. J Clin Endocrinol Metab. 2025;110(4):911‐921. [DOI] [PubMed] [Google Scholar]
- 13. Bowen RL, Smith MA, Harris PLR, et al. Elevated luteinizing hormone expression colocalizes with neurons vulnerable to Alzheimer's disease pathology. J Neurosci Res. 2002;70(3):514‐518. [DOI] [PubMed] [Google Scholar]
- 14. Ipsa E, Cruzat VF, Kagize JN, Yovich JL, Keane KN. Growth hormone and insulin-like growth factor action in reproductive tissues. Front Endocrinol (Lausanne). 2019;10:777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Kauffman AS. Neuroendocrine mechanisms underlying estrogen positive feedback and the LH surge. Front Neurosci. 2022;16:953252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Egbert JR, Fahey PG, Reimer J, et al. Follicle-stimulating hormone and luteinizing hormone increase Ca2 + in the granulosa cells of mouse ovarian follicles†. Biol Reprod. 2019;101(2):433‐444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Blair JA, McGee H, Bhatta S, Palm R, Casadesus G. Hypothalamic-pituitary-gonadal axis involvement in learning and memory and Alzheimer's disease: more than “just” estrogen. Front Endocrinol (Lausanne). 2015;6:45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Sela-Abramovich S, Chorev E, Galiani D, Dekel N. Mitogen-activated protein kinase mediates luteinizing hormone-induced breakdown of communication and oocyte maturation in rat ovarian follicles. Endocrinology. 2005;146(3):1236‐1244. [DOI] [PubMed] [Google Scholar]
- 19. Reed BG, Carr BR. The normal menstrual cycle and the control of ovulation. Endotext. Published online August 5, 2018. Accessed June 9, 2025. https://www.ncbi.nlm.nih.gov/books/NBK279054/
- 20. Holschbach MA, Handa RJ. Androgen action and stress. Stress: Neuroendocrinol and Neurobiol. 2017;2:227‐235. [Google Scholar]
- 21. Kauffman AS. Androgen inhibition of reproductive neuroendocrine function in females and transgender males. Endocrinology. 2024;165(10):bqae113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Halvorson LM, DeCherney AH. Inhibin, activin, and follistatin in reproductive medicine. Fertil Steril. 1996;65(3):459‐469. [DOI] [PubMed] [Google Scholar]
- 23. Alam MH, Miyano T. Interaction between growing oocytes and granulosa cells in vitro. Reprod Med Biol. 2020;19(1):13‐23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Peacock K, Carlson K, Ketvertis KM. Menopause. In: StatPearls [Internet]; [Updated December 21, 2023]. StatPearls Publishing; 2025. Accessed June 9, 2025. https://www.ncbi.nlm.nih.gov/books/NBK507826/ [Google Scholar]
- 25. Gold EB, Crawford SL, Avis NE, et al. Factors related to age at natural menopause: longitudinal analyses from SWAN. Am J Epidemiol. 2013;178(1):70‐83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Stankovic S, Shekari S, Huang QQ, et al. Genetic links between ovarian ageing, cancer risk and de novo mutation rates. Nature. 2024;633(8030):608‐614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Li M, Zhu Y, Wei J, Chen L, Chen S, Lai D. The global prevalence of premature ovarian insufficiency: a systematic review and meta-analysis. Climacteric. 2023;26(2):95‐102. [DOI] [PubMed] [Google Scholar]
- 28. Sochocka M, Karska J, Pszczołowska M, et al. Cognitive decline in early and premature menopause. Int J Mol Sci. 2023;24(7):6566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Fortuño C, Labarta E. Genetics of primary ovarian insufficiency: a review. J Assist Reprod Genet. 2014;31(12):1573‐1585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Yang J, Chen C. Hormonal changes in PCOS. J Endocrinol. 2024;261(1):e230342. [DOI] [PubMed] [Google Scholar]
- 31. Dinsdale NL, Crespi BJ. Endometriosis and polycystic ovary syndrome are diametric disorders. Evol Appl. 2021;14(7):1693‐1715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Khan MJ, Ullah A, Basit S. Genetic basis of polycystic ovary syndrome (PCOS): current perspectives. Appl Clin Genet. 2019;12:249‐260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Tian Y, Li J, Su S, et al. PCOS-GWAS susceptibility variants in THADA, INSR, TOX3, and DENND1A are associated with metabolic syndrome or insulin resistance in women with PCOS. Front Endocrinol (Lausanne). 2020;11:274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. McAllister JM, Modi B, Miller BA, et al. Overexpression of a DENND1A isoform produces a polycystic ovary syndrome theca phenotype. Proc Natl Acad Sci U S A. 2014;111(15):E1519‐E1527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Rahmioglu N, Mortlock S, Ghiasi M, et al. The genetic basis of endometriosis and comorbidity with other pain and inflammatory conditions. Nat Genet. 2023;55(3):423‐436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Gao Y, Zou Y, Wu G, Zheng L. Oxidative stress and mitochondrial dysfunction of granulosa cells in polycystic ovarian syndrome. Front Med (Lausanne). 2023;10:1193749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Fan W, Yuan Z, Li M, Zhang Y, Nan F. Decreased oocyte quality in patients with endometriosis is closely related to abnormal granulosa cells. Front Endocrinol (Lausanne). 2023;14:1226687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Shang J, Wang S, Wang A, et al. Intra-ovarian inflammatory states and their associations with embryo quality in normal-BMI PCOS patients undergoing IVF treatment. Reprod Biol Endocrinol. 2024;22(1):11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Correa DD, Zhou Q, Thaler HT, Maziarz M, Hurley K, Hensley ML. Cognitive functions in long-term survivors of ovarian cancer. Gynecol Oncol. 2010;119(2):366‐369. [DOI] [PubMed] [Google Scholar]
- 40. Chantalat E, Valera M-C, Vaysse C, et al. Estrogen receptors and endometriosis. Int J Mol Sci. 2020;21(8):2815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Wang T-M, Lee Y-L, Chung C-H, et al. Association between endometriosis and mental disorders including psychiatric disorders, suicide, and all-cause mortality -a nationwide population-based cohort study in Taiwan. Int J Womens Health. 2023;15:1865‐1882. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Richmond-Rakerd LS, D'Souza S, Milne BJ, Caspi A, Moffitt TE. Longitudinal associations of mental disorders with dementia. JAMA Psychiatry. 2022;79(4):333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Maulitz L, Stickeler E, Stickel S, Habel U, Tchaikovski SN, Chechko N. Endometriosis, psychiatric comorbidities and neuroimaging: estimating the odds of an endometriosis brain. Front Neuroendocrinol. 2022;65:100988. [DOI] [PubMed] [Google Scholar]
- 44. Mosconi L, Berti V, Dyke J, et al. Menopause impacts human brain structure, connectivity, energy metabolism, and amyloid-beta deposition. Sci Rep. 2021;11(1):10867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. 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] [PMC free article] [PubMed] [Google Scholar]
- 46. Christensen A, Pike CJ. Menopause, obesity and inflammation: interactive risk factors for Alzheimer's disease. Front Aging Neurosci. 2015;7:130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Yonker JA, Chang V, Roetker NS, Hauser TS, Hauser RM, Atwood CS. Hypothalamic-pituitary-gonadal axis homeostasis predicts longevity. Age (Dordr). 2013;35(1):129‐138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Malek AM, Vladutiu CJ, Meyer ML, et al. The association of age at menopause and all-cause and cause-specific mortality by race, postmenopausal hormone use, and smoking status. Prev Med Rep. 2019;15:100955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Moolhuijsen LME, Visser JA. Anti-Müllerian hormone and ovarian reserve: update on assessing ovarian function. J Clin Endocrinol Metab. 2020;105(11):3361‐3373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Vujović S, Ivović M, Gajić MT, et al. Alzheimer's disease and premature ovarian insufficiency. Endocrines. 2023;4(2):250‐256. [Google Scholar]
- 51. Xi H, Gan J, Liu S, et al. Reproductive factors and cognitive impairment in natural menopausal women: a cross-sectional study. Front Endocrinol (Lausanne). 2022;13:893901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Jahanfar S, Eden JA, Warren P, Seppälä M, Nguyen TV. A twin study of polycystic ovary syndrome. Fertil Steril. 1995;63(3):478‐486. [PubMed] [Google Scholar]
- 53. Vine D, Ghosh M, Wang T, Bakal J. Increased prevalence of adverse health outcomes across the lifespan in those affected by polycystic ovary syndrome: a Canadian population cohort. CJC Open. 2024;6(2Part B):314‐326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Wilson DM, Cookson MR, Van Den et al. Hallmarks of neurodegenerative diseases. Cell. 2023;186(4):693‐714. [DOI] [PubMed] [Google Scholar]
- 55. Ahmad MH, Fatima M, Mondal AC. Role of hypothalamic-pituitary-adrenal axis, hypothalamic-pituitary-gonadal axis and insulin signaling in the pathophysiology of Alzheimer's disease. Neuropsychobiology. 2019;77(4):197‐205. [DOI] [PubMed] [Google Scholar]
- 56. Bousfield GR, Dias JA. Synthesis and secretion of gonadotropins including structure-function correlates. Rev Endocr Metab Disord. 2011;12(4):289‐302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Chagnot A, Montagne A. Connexins and blood-brain barrier: beyond the gap. Neuron. 2023;111(22):3499‐3501. [DOI] [PubMed] [Google Scholar]
- 58. Manu DR, Slevin M, Barcutean L, Forro T, Boghitoiu T, Balasa R. Astrocyte involvement in blood-brain barrier function: a critical update highlighting novel, complex, neurovascular interactions. Int J Mol Sci. 2023;24(24):17146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Banwinkler M, Theis H, Prange S, van Eimeren T. Imaging the limbic system in Parkinson's disease-a review of limbic pathology and clinical symptoms. Brain Sci. 2022;12(9):1248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Fjell AM, Westlye LT, Grydeland H, et al. Critical ages in the life course of the adult brain: nonlinear subcortical aging. Neurobiol Aging. 2013;34(10):2239‐2247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Zhang H, Roman RJ, Fan F. Hippocampus is more susceptible to hypoxic injury: has the Rosetta Stone of regional variation in neurovascular coupling been deciphered? Geroscience. 2022;44(1):127‐130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Kapogiannis D, Mattson MP. Disrupted energy metabolism and neuronal circuit dysfunction in cognitive impairment and Alzheimer's disease. Lancet Neurol. 2011;10(2):187‐198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Jin K, Yao Z, van Velthoven CTJ, et al. Brain-wide cell-type-specific transcriptomic signatures of healthy ageing in mice. Nature. 2025;638(8049):182‐196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Boisvert MM, Erikson GA, Shokhirev MN, Allen NJ. The aging astrocyte transcriptome from multiple regions of the mouse brain. Cell Rep. 2018;22(1):269‐285. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Vercruysse P, Vieau D, Blum D, Petersén Å, Dupuis L. Hypothalamic alterations in neurodegenerative diseases and their relation to abnormal energy metabolism. Front Mol Neurosci. 2018;11:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Hensler JG. Serotonergic modulation of the limbic system. Neurosci Biobehav Rev. 2006;30(2):203‐214. [DOI] [PubMed] [Google Scholar]
- 67. Hara Y, Yuk F, Puri R, Janssen WGM, Rapp PR, Morrison JH. Presynaptic mitochondrial morphology in monkey prefrontal cortex correlates with working memory and is improved with estrogen treatment. Proc Natl Acad Sci U S A. 2014;111(1):486‐491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Xiong J, Kang SS, Wang Z, et al. FSH blockade improves cognition in mice with Alzheimer's disease. Nature. 2022;603(7901):470‐476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Brann DW, Lu Y, Wang J, et al. Brain-derived estrogen and neural function. Neurosci Biobehav Rev. 2022;132:793‐817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Lu Y, Sareddy GR, Wang J, et al. Neuron-derived estrogen regulates synaptic plasticity and memory. J Neurosci. 2019;39(15):2792‐2809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Yao M, Nguyen T-VV, Pike CJ. Estrogen regulates Bcl-w and Bim expression: role in protection against beta-amyloid peptide-induced neuronal death. J Neurosci. 2007;27(6):1422‐1433. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Nyúl-Tóth Á, Patai R, Csiszar A, et al. Linking peripheral atherosclerosis to blood-brain barrier disruption: elucidating its role as a manifestation of cerebral small vessel disease in vascular cognitive impairment. Geroscience. 2024;46(6):6511‐6536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Zhao Y, Zhang X, Chen X, Wei Y. Neuronal injuries in cerebral infarction and ischemic stroke: from mechanisms to treatment (Review). Int J Mol Med. 2022;49(2):15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Xiao Z-M, Sun L, Liu Y-M, Zhang J-J, Huang J. Estrogen regulation of the neprilysin gene through a hormone-responsive element. J Mol Neurosci. 2009;39(1-2):22‐26. [DOI] [PubMed] [Google Scholar]
- 75. Suzuki S, Brown CM, Wise PM. Neuroprotective effects of estrogens following ischemic stroke. Front Neuroendocrinol. 2009;30(2):201‐211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. McArthur S, Loiola RA, Maggioli E, Errede M, Virgintino D, Solito E. The restorative role of annexin A1 at the blood-brain barrier. Fluids Barriers CNS. 2016;13(1):17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Loiola RA, Wickstead ES, Solito E, McArthur S. Estrogen promotes pro-resolving microglial behavior and phagocytic cell clearance through the actions of annexin A1. Front Endocrinol (Lausanne). 2019;10:420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Baudry M, Bi X, Aguirre C. Progesterone-estrogen interactions in synaptic plasticity and neuroprotection. Neuroscience. 2013;239:280‐294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Noorbakhsh F, Baker GB, Power C. Allopregnanolone and neuroinflammation: a focus on multiple sclerosis. Front Cell Neurosci. 2014;8:134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. De Nicola AF, Gonzalez Deniselle MC, Garay L, et al. Progesterone protective effects in neurodegeneration and neuroinflammation. J Neuroendocrinol. 2013;25(11):1095‐1103. [DOI] [PubMed] [Google Scholar]
- 81. Yilmaz C, Karali K, Fodelianaki G, et al. Neurosteroids as regulators of neuroinflammation. Front Neuroendocrinol. 2019;55:100788. [DOI] [PubMed] [Google Scholar]
- 82. Ishrat T, Sayeed I, Atif F, Hua F, Stein DG. Progesterone and allopregnanolone attenuate blood-brain barrier dysfunction following permanent focal ischemia by regulating the expression of matrix metalloproteinases. Exp Neurol. 2010;226(1):183‐190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Ishihara Y, Fujitani N, Sakurai H, et al. Effects of sex steroid hormones and their metabolites on neuronal injury caused by oxygen-glucose deprivation/reoxygenation in organotypic hippocampal slice cultures. Steroids. 2016;113:71‐77. [DOI] [PubMed] [Google Scholar]
- 84. Cai Y, Chew C, Muñoz F, Sengelaub DR. Neuroprotective effects of testosterone metabolites and dependency on receptor action on the morphology of somatic motoneurons following the death of neighboring motoneurons. Dev Neurobiol. 2017;77(6):691‐707. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Colciago A, Casati L, Negri-Cesi P, Celotti F. Learning and memory: steroids and epigenetics. J Steroid Biochem Mol Biol. 2015;150:64‐85. [DOI] [PubMed] [Google Scholar]
- 86. Li M, Masugi-Tokita M, Takanami K, Yamada S, Kawata M. Testosterone has sublayer-specific effects on dendritic spine maturation mediated by BDNF and PSD-95 in pyramidal neurons in the hippocampus CA1 area. Brain Res. 2012;1484:76‐84. [DOI] [PubMed] [Google Scholar]
- 87. McAllister C, Long J, Bowers A, et al. Genetic targeting aromatase in male amyloid precursor protein transgenic mice down-regulates beta-secretase (BACE1) and prevents Alzheimer-like pathology and cognitive impairment. J Neurosci. 2010;30(21):7326‐7334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Yu Z, Jiao Y, Zhao Y, Gu W. Level of estrogen in females—the different impacts at different life stages. J Pers Med. 2022;12(12):1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Schulster M, Bernie A, Ramasamy R. The role of estradiol in male reproductive function. Asian J Androl. 2016;18(3):435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Manly JJ, Merchant CA, Jacobs DM, et al. Endogenous estrogen levels and Alzheimer's disease among postmenopausal women. Neurology. 2000;54(4):833‐837. [DOI] [PubMed] [Google Scholar]
- 91. Yao J, Chen S, Cadenas E, Brinton RD. Estrogen protection against mitochondrial toxin-induced cell death in hippocampal neurons: antagonism by progesterone. Brain Res. 2011;1379:2‐10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Schattmann L, Sherwin BB. Testosterone levels and cognitive functioning in women with polycystic ovary syndrome and in healthy young women. Horm Behav. 2007;51(5):587‐596. [DOI] [PubMed] [Google Scholar]
- 93. Nierwińska K, Nowacka-Chmielewska M, Bernacki J, et al. The effect of endurance training and testosterone supplementation on the expression of blood spinal cord barrier proteins in rats. PLoS One. 2019;14(2):e0211818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Atallah A, Mhaouty-Kodja S, Grange-Messent V. Chronic depletion of gonadal testosterone leads to blood-brain barrier dysfunction and inflammation in male mice. J Cereb Blood Flow Metab. 2017;37(9):3161‐3175. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Huang L, Sun S, Jiang G, et al. Follicle-stimulating hormone induces depression-like phenotype by affecting synaptic function. Front Mol Neurosci. 2024;17:1459858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Xiong J, Kang SS, Wang M, et al. FSH and ApoE4 contribute to Alzheimer's disease-like pathogenesis via C/EBPβ/δ-secretase in female mice. Nat Commun. 2023;14(1):6577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Hyde Z, Flicker L, Almeida OP, et al. Higher luteinizing hormone is associated with poor memory recall: the health in men study. J Alzheimers Dis. 2010;19(3):943‐951. [DOI] [PubMed] [Google Scholar]
- 98. Bowen RL, Verdile G, Liu T, et al. Luteinizing hormone, a reproductive regulator that modulates the processing of amyloid-beta precursor protein and amyloid-beta deposition. J Biol Chem. 2004;279(19):20539‐20545. [DOI] [PubMed] [Google Scholar]
- 99. Burnham VL, Thornton JE. Luteinizing hormone as a key player in the cognitive decline of Alzheimer's disease. Horm Behav. 2015;76:48‐56. [DOI] [PubMed] [Google Scholar]
- 100. Berry A, Tomidokoro Y, Ghiso J, Thornton J. Human chorionic gonadotropin (a luteinizing hormone homologue) decreases spatial memory and increases brain amyloid-beta levels in female rats. Horm Behav. 2008;54(1):143‐152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Liu T, Wimalasena J, Bowen RL, Atwood CS. Luteinizing hormone receptor mediates neuronal pregnenolone production via up-regulation of steroidogenic acute regulatory protein expression. J Neurochem. 2007;100(5):1329‐1339. [DOI] [PubMed] [Google Scholar]
- 102. Casadesus G, Webber KM, Atwood CS, et al. Luteinizing hormone modulates cognition and amyloid-beta deposition in Alzheimer APP transgenic mice. Biochim Biophys Acta. 2006;1762(4):447‐452. [DOI] [PubMed] [Google Scholar]
- 103. Bohm-Levine N, Goldberg AR, Mariani M, Frankfurt M, Thornton J. Reducing luteinizing hormone levels after ovariectomy improves spatial memory: possible role of brain-derived neurotrophic factor. Horm Behav. 2020;118:104590. [DOI] [PubMed] [Google Scholar]
- 104. Wilson AC, Clemente L, Liu T, Bowen RL, Meethal SV, Atwood CS. Reproductive hormones regulate the selective permeability of the blood-brain barrier. Biochim Biophys Acta. 2008;1782(6):401‐407. [DOI] [PubMed] [Google Scholar]
- 105. Mey M, Bhatta S, Casadesus G. Luteinizing hormone and the aging brain. Vitam Horm. 2021;115:89‐104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Short RA, O’Brien PC, Graff-Radford NR, Bowen RL. Elevated gonadotropin levels in patients with Alzheimer disease. Mayo Clin Proc. 2001;76(9):906‐909. [DOI] [PubMed] [Google Scholar]
- 107. Bowen RL, Isley JP, Atkinson RL. An association of elevated serum gonadotropin concentrations and Alzheimer disease? J Neuroendocrinol. 2000;12(4):351‐354. [DOI] [PubMed] [Google Scholar]
- 108. Oliver C, Mical RS, Porter JC. Hypothalamic-pituitary vasculature: evidence for retrograde blood flow in the pituitary stalk 1. Endocrinology. 1977;101(2):598‐604. [DOI] [PubMed] [Google Scholar]
- 109. Lukacs H, Hiatt ES, Lei ZM, Rao C. Peripheral and intracerebroventricular administration of human chorionic gonadotropin alters several hippocampus-associated behaviors in cycling female rats. Horm Behav. 1995;29(1):42‐58. [DOI] [PubMed] [Google Scholar]
- 110. Rodrigues MA, Verdile G, Foster JK, et al. Gonadotropins and cognition in older women. J Alzheimer's Dis. 2008;13(3):267‐274. [DOI] [PubMed] [Google Scholar]
- 111. Casadesus G, Milliken EL, Webber KM, et al. Increases in luteinizing hormone are associated with declines in cognitive performance. Mol Cell Endocrinol. 2007;269(1-2):107‐111. [DOI] [PubMed] [Google Scholar]
- 112. Burnham V, Sundby C, Laman-Maharg A, Thornton J. Luteinizing hormone acts at the hippocampus to dampen spatial memory. Horm Behav. 2017;89:55‐63. [DOI] [PubMed] [Google Scholar]
- 113. Blair JA, Bhatta S, Casadesus G. CNS luteinizing hormone receptor activation rescues ovariectomy-related loss of spatial memory and neuronal plasticity. Neurobiol Aging. 2019;78:111‐120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Emanuele N, Oslapas R, Connick E, Kirsteins L, Lawrence AM. Hypothalamic LH may play a role in control of pituitary LH release. Neuroendocrinology. 1981;33(1):12‐17. [DOI] [PubMed] [Google Scholar]
- 115. Peegel H, Randolph J, Midgley AR, Menon KM. In situ hybridization of luteinizing hormone/human chorionic gonadotropin receptor messenger ribonucleic acid during hormone-induced down-regulation and the subsequent recovery in rat corpus luteum. Endocrinology. 1994;135(3):1044‐1051. [DOI] [PubMed] [Google Scholar]
- 116. Lei ZM, Rao CV, Kornyei JL, Licht P, Hiatt ES. Novel expression of human chorionic gonadotropin/luteinizing hormone receptor gene in brain. Endocrinology. 1993;132(5):2262‐2270. [DOI] [PubMed] [Google Scholar]
- 117. Hebert AE, Dash PK. Extracellular signal-regulated kinase activity in the entorhinal cortex is necessary for long-term spatial memory. Learning & Memory. 2002;9(4):156‐166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Wenner MM, Shenouda N, Shoemaker L, et al. Characterizing vascular and hormonal changes in women across the life span: a cross-sectional analysis. Am J Physiol Heart Circ Physiol. 2024;327(5):H1286‐H1295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Abildgaard J, Tingstedt J, Zhao Y, et al. Increased systemic inflammation and altered distribution of T-cell subsets in postmenopausal women. PLoS One. 2020;15(6):e0235174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Ramachandran Pillai R, Sharon L, Premkumar NR, Kattimani S, Sagili H, Rajendiran S. Luteinizing hormone-follicle stimulating hormone ratio as biological predictor of post-partum depression. Compr Psychiatry. 2017;72:25‐33. [DOI] [PubMed] [Google Scholar]
- 121. Harlow BL, Wise LA, Otto MW, Soares CN, Cohen LS. Depression and its influence on reproductive endocrine and menstrual cycle markers associated with perimenopause: the Harvard study of moods and cycles. Arch Gen Psychiatry. 2003;60(1):29‐36. [DOI] [PubMed] [Google Scholar]
- 122. Ryu V, Gumerova A, Korkmaz F, et al. Brain atlas for glycoprotein hormone receptors at single-transcript level. Elife. 2022:11:e79612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Huo S-D, Chen S-E, Long R-J, et al. Protein and mRNA expression of follicle-stimulating hormone receptor and luteinizing hormone receptor during the oestrus in the yak (Bos grunniens). Reprod Domest Anim. 2017;52(3):477‐482. [DOI] [PubMed] [Google Scholar]
- 124. Bi W-K, Luan S-S, Wang J, et al. FSH signaling is involved in affective disorders. Biochem Biophys Res Commun. 2020;525(4):915‐920. [DOI] [PubMed] [Google Scholar]
- 125. Qian H, Jia J, Yang Y, Bian Z, Ji Y. A follicle-stimulating hormone exacerbates the progression of periapical inflammation through modulating the cytokine release in periodontal tissue. Inflammation. 2020;43(4):1572‐1585. [DOI] [PubMed] [Google Scholar]
- 126. Chen Y, Xu N, Zhang W, et al. FSH enhances the inflammatory response of macrophages in the knee joint possibly through the NFκB pathway. FEBS Open Bio. 2025;15(4):622‐633. [Google Scholar]
- 127. Xue Y, Zuo S, Wang F, Qi X. From hormones to neurodegeneration: how FSH drives Alzheimer's disease. Front Aging Neurosci. 2025;17:1578439. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Drevets WC, Price JL, Furey ML. Brain structural and functional abnormalities in mood disorders: implications for neurocircuitry models of depression. Brain Struct Funct. 2008;213(1-2):93‐118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Van Someren EJW. Brain mechanisms of insomnia: new perspectives on causes and consequences. Physiol Rev. 2021;101(3):995‐1046. [DOI] [PubMed] [Google Scholar]
- 130. Pandya M, Altinay M, Malone DA, Anand A. Where in the brain is depression? Curr Psychiatry Rep. 2012;14(6):634‐642. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Price JL, Drevets WC. Neurocircuitry of mood disorders. Neuropsychopharmacology. 2010;35(1):192‐216. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Freedman RR, Benton MD, Genik RJ, Graydon FX. Cortical activation during menopausal hot flashes. Fertil Steril. 2006;85(3):674‐678. [DOI] [PubMed] [Google Scholar]
- 133. Padilla SL, Johnson CW, Barker FD, Patterson MA, Palmiter RD. A neural circuit underlying the generation of hot flushes. Cell Rep. 2018;24(2):271‐277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Thurston RC, Maki PM, Derby CA, Sejdić E, Aizenstein HJ. Menopausal hot flashes and the default mode network. Fertil Steril. 2015;103(6):1572‐1578.e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Diwadkar VA, Murphy ER, Freedman RR. Temporal sequencing of brain activations during naturally occurring thermoregulatory events. Cereb Cortex. 2014;24(11):3006‐3013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Helquist K, Baughan E, Pouwer L, et al. Causal brain network implicated in insomnia symptoms (S36.006). Neurology. 2024;102(17_Suppl_1):6455‐6455. [Google Scholar]
- 137. Schiel JE, Holub F, Petri R, et al. Affect and arousal in insomnia: through a Lens of neuroimaging studies. Curr Psychiatry Rep. 2020;22(9):44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Osorio-Forero A, Foustoukos G, Cardis R, et al. Infraslow noradrenergic locus coeruleus activity fluctuations are gatekeepers of the NREM-REM sleep cycle. Nat Neurosci. 2025;28(1):84‐96. [DOI] [PubMed] [Google Scholar]
- 139. Levenson JC, Kay DB, Buysse DJ. The pathophysiology of insomnia. Chest. 2015;147(4):1179‐1192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Hugon J, Msika EF, Queneau M, Farid K, Paquet C. Long COVID: cognitive complaints (brain fog) and dysfunction of the cingulate cortex. J Neurol. 2022;269(1):44‐46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Díez-Cirarda M, Yus M, Gómez-Ruiz N, et al. Multimodal neuroimaging in post-COVID syndrome and correlation with cognition. Brain. 2023;146(5):2142‐2152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Denno P, Zhao S, Husain M, Hampshire A. Defining brain fog across medical conditions. Trends Neurosci. 2025;48(5):330‐348. [DOI] [PubMed] [Google Scholar]
- 143. Pan Y, Hu J. Hormonal basis of brain fog in cancer treatment. J Exp Med. 2024;221(5):e20231942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Schupf N, Winsten S, Patel B, et al. Bioavailable estradiol and age at onset of Alzheimer's disease in postmenopausal women with Down syndrome. Neurosci Lett. 2006;406(3):298‐302. [DOI] [PubMed] [Google Scholar]
- 145. Biegon A, McEwen BS. Modulation by estradiol of serotonin receptors in brain. J Neurosci. 1982;2(2):199‐205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Molendijk ML, Bus BAA, Spinhoven P, et al. Serum levels of brain-derived neurotrophic factor in major depressive disorder: state-trait issues, clinical features and pharmacological treatment. Mol Psychiatry. 2011;16(11):1088‐1095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Verdile G, Yeap BB, Clarnette RM, et al. Luteinizing hormone levels are positively correlated with plasma amyloid-beta protein levels in elderly men. J Alzheimers Dis. 2008;14(2):201‐208. [DOI] [PubMed] [Google Scholar]
- 148. Nerattini M, Rubino F, Jett S, et al. Elevated gonadotropin levels are associated with increased biomarker risk of Alzheimer's disease in midlife women. Front Dement. 2023;2:1303256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Spicer J, Malaspina D, Blank SV, Goosens KA. Follicle-stimulating hormone: more than a marker for menopause: FSH as a frontier for women's mental health. Psychiatry Res. 2025;345:116239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Arfa-Fatollahkhani P, Nahavandi A, Abtahi H, et al. The effect of luteinizing hormone reducing agent on anxiety and novel object recognition memory in gonadectomized rats. Basic Clin Neurosci. 2017;8(2):113‐119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Talbot K, Wang HY, Kazi H, et al. Demonstrated brain insulin resistance in Alzheimer's disease patients is associated with IGF-1 resistance, IRS-1 dysregulation, and cognitive decline. J Clin Invest. 2012;122(4):1316‐1338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Wu L, Qin Y, Yuan H, Zhu Y, Hu A. Anti-inflammatory and neuroprotective effects of insulin-like growth factor-1 overexpression in pentylenetetrazole (PTZ)-induced mouse model of chronic epilepsy. Brain Res. 2022;1785:147881. [DOI] [PubMed] [Google Scholar]
- 153. Kellar D, Craft S. Brain insulin resistance in Alzheimer's disease and related disorders: mechanisms and therapeutic approaches. Lancet Neurol. 2020;19(9):758‐766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Sarahian N, Sarvazad H, Sajadi E, Rahnejat N, Eskandari Roozbahani N. Investigation of common risk factors between polycystic ovary syndrome and Alzheimer's disease: a narrative review. Reprod Health. 2021;18(1):156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Pedersen DJ, Guilherme A, Danai LV, et al. A major role of insulin in promoting obesity-associated adipose tissue inflammation. Mol Metab. 2015;4(7):507‐518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Zhang B, Zhang Y, Wu W, et al. Chronic glucocorticoid exposure activates BK-NLRP1 signal involving in hippocampal neuron damage. J Neuroinflammation. 2017;14(1):139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Popp J, Wolfsgruber S, Heuser I, et al. Cerebrospinal fluid cortisol and clinical disease progression in MCI and dementia of Alzheimer's type. Neurobiol Aging. 2015;36(2):601‐607. [DOI] [PubMed] [Google Scholar]
- 158. Green KN, Billings LM, Roozendaal B, McGaugh JL, LaFerla FM. Glucocorticoids increase amyloid-beta and tau pathology in a mouse model of Alzheimer's disease. J Neurosci. 2006;26(35):9047‐9056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Kunicki M, Rzewuska N, Gross-Kępińska K. Immunophenotypic profiles and inflammatory markers in premature ovarian insufficiency. J Reprod Immunol. 2024;164:104253. [DOI] [PubMed] [Google Scholar]
- 160. Malutan AM, Dan M, Nicolae C, Carmen M. Proinflammatory and anti-inflammatory cytokine changes related to menopause. Prz Menopauzalny. 2014;13(3):162‐168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Farkas S, Szabó A, Török B, et al. Ovariectomy-induced hormone deprivation aggravates Aβ1-42 deposition in the basolateral amygdala and cholinergic fiber loss in the cortex but not cognitive behavioral symptoms in a triple transgenic mouse model of Alzheimer's disease. Front Endocrinol (Lausanne). 2022;13:985424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Van Goozen SH, Cohen-Kettenis PT, Gooren LJ, Frijda NH, Van de Poll NE. Activating effects of androgens on cognitive performance: causal evidence in a group of female-to-male transsexuals. Neuropsychologia. 1994;32(10):1153‐1157. [DOI] [PubMed] [Google Scholar]
- 163. Perović M, Wugalter K, Einstein G. Review of the effects of polycystic ovary syndrome on cognition: looking beyond the androgen hypothesis. Front Neuroendocrinol. 2022;67:101038. [DOI] [PubMed] [Google Scholar]
- 164. Krishnan A, Muthusami S, Periyasamy L, Stanley JA, Gopalakrishnan V, Ramachandran I. Effect of DHT-induced hyperandrogenism on the pro-inflammatory cytokines in a rat model of polycystic ovary morphology. Medicina (Kaunas). 2020;56(3):100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Zhao H, Zhang J, Cheng X, Nie X, He B. Insulin resistance in polycystic ovary syndrome across various tissues: an updated review of pathogenesis, evaluation, and treatment. J Ovarian Res. 2023;16(1):9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Musial N, Ali Z, Grbevski J, Veerakumar A, Sharma P. Perimenopause and first-onset mood disorders: a closer look. Focus (Am Psychiatr Publ). 2021;19(3):330‐337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Copinschi G, Caufriez A. Sleep and the ovarian axis. Curr Opin Endocr Metab Res. 2021;17:38‐45. [Google Scholar]
- 168. Berga SL. Brain phenotype in PCOS: androgens, anovulation, and gender. Gynecol Endocrinol. 2022;38(8):615‐616. [DOI] [PubMed] [Google Scholar]
- 169. Brutocao C, Zaiem F, Alsawas M, Morrow AS, Murad MH, Javed A. Psychiatric disorders in women with polycystic ovary syndrome: a systematic review and meta-analysis. Endocrine. 2018;62(2):318‐325. [DOI] [PubMed] [Google Scholar]
- 170. Flores-Ramos M, Martínez-Mota L, Silvestri-Tommasoni R, Jiménez Domínguez G, Linares Silva D. Free testosterone is associated with perceived stress in women. Pers Med Psychiatry. 2024;45-46:100129. [Google Scholar]
- 171. Joseph JJ, Golden SH. Cortisol dysregulation: the bidirectional link between stress, depression, and type 2 diabetes mellitus. Ann N Y Acad Sci. 2017;1391(1):20‐34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Cui P, Hu W, Ma T, et al. Long-term androgen excess induces insulin resistance and non-alcoholic fatty liver disease in PCOS-like rats. J Steroid Biochem Mol Biol. 2021;208:105829. [DOI] [PubMed] [Google Scholar]
- 173. González F. Inflammation in polycystic ovary syndrome: underpinning of insulin resistance and ovarian dysfunction. Steroids. 2012;77(4):300‐305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Beral V, Bull D, Reeves G, Million Women Study Collaborators . Endometrial cancer and hormone-replacement therapy in the million women study. Lancet. 2005;365(9470):1543‐1551. [DOI] [PubMed] [Google Scholar]
- 175. Roseweir AK, Millar RP. Kisspeptin antagonists. Adv Exp Med Biol. 2013;784:159‐186. [DOI] [PubMed] [Google Scholar]
- 176. Lederman S, Ottery FD, Cano A, et al. Fezolinetant for treatment of moderate-to-severe vasomotor symptoms associated with menopause (SKYLIGHT 1): a phase 3 randomised controlled study. Lancet. 2023;401(10382):1091‐1102. [DOI] [PubMed] [Google Scholar]
- 177. Landry DA, Yakubovich E, Cook DP, Fasih S, Upham J, Vanderhyden BC. Metformin prevents age-associated ovarian fibrosis by modulating the immune landscape in female mice. Sci Adv. 2022;8(35):eabq1475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178. Simpson AMR, De Souza MJ, Damani J, et al. Gut microbes differ in postmenopausal women responding to prunes to maintain hip bone mineral density. Front Nutr. 2024;11:1389638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Meslier V, Laiola M, Roager HM, et al. Mediterranean diet intervention in overweight and obese subjects lowers plasma cholesterol and causes changes in the gut microbiome and metabolome independently of energy intake. Gut. 2020;69(7):1258‐1268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Chen Y, Feng R, Yang X, et al. Yogurt improves insulin resistance and liver fat in obese women with nonalcoholic fatty liver disease and metabolic syndrome: a randomized controlled trial. Am J Clin Nutr. 2019;109(6):1611‐1619. [DOI] [PubMed] [Google Scholar]
- 181. Thevaranjan N, Puchta A, Schulz C, et al. Age-associated microbial dysbiosis promotes intestinal permeability, systemic inflammation, and macrophage dysfunction. Cell Host Microbe. 2017;21(4):455‐466.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Ghosh TS, Shanahan F, O’Toole PW. The gut microbiome as a modulator of healthy ageing. Nat Rev Gastroenterol Hepatol. 2022;19(9):565‐584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183. Son WH, Park HT, Jeon BH, Ha MS. Moderate intensity walking exercises reduce the body mass index and vascular inflammatory factors in postmenopausal women with obesity: a randomized controlled trial. Sci Rep. 2023;13(1):20172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Pandit U, Singh M, Ranjan R, Gupta V. The effect of exercise training on body composition, insulin resistance and high sensitivity C-reactive protein (Hs-CRP) in women with polycystic ovary syndrome: a pilot study from north India. Cureus. 2022;14(4):e23994. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Data sharing is not applicable to this article as no data sets were generated or analyzed during the current study.