Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 74
1.
J Appl Microbiol ; 135(4)2024 Apr 01.
Article En | MEDLINE | ID: mdl-38253409

AIMS: To examine the influence of GED on the gut microbiota and metabolites using a bilateral ovariectomized (OVX) rat model. We tried to elucidate the underlying mechanisms of GED in the treatment of menopausal hot flashes. METHODS AND RESULTS: 16S rRNA sequencing, metabonomics, molecular biological analysis, and fecal microbiota transplantation (FMT) were conducted to elucidate the mechanisms by which GED regulates the gut microbiota. GED significantly reduced OVX-induced hot flashes and improved disturbances in the gut microbiota metabolites. Moreover, FMT validated that the gut microbiota can trigger hot flashes, while GED can alleviate hot flash symptoms by modulating the composition of the gut microbiota. Specifically, GED upregulated the abundance of Blautia, thereby increasing l(+)-ornithine levels for the treatment of menopausal hot flashes. Additionally, GED affected endothelial nitric oxide synthase and heat shock protein 70 (HSP70) levels in the hypothalamic preoptic area by changing the gut microbiota composition. CONCLUSIONS: Our study illuminated the underlying mechanisms by which GED attenuated the hot flashes through modulation of the gut microbiota and explored the regulatory role of the gut microbiota on HSP70 expression in the preoptic anterior hypothalamus, thereby establishing a foundation for further exploration of the role of the gut-brain axis in hot flashes.


Drugs, Chinese Herbal , Gastrointestinal Microbiome , Hot Flashes , Menopause , Animals , Gastrointestinal Microbiome/drug effects , Hot Flashes/metabolism , Hot Flashes/drug therapy , Rats , Female , Drugs, Chinese Herbal/pharmacology , Fecal Microbiota Transplantation , Ovariectomy , Rats, Sprague-Dawley , RNA, Ribosomal, 16S/genetics , Metabolome/drug effects
2.
Best Pract Res Clin Endocrinol Metab ; 38(1): 101774, 2024 Jan.
Article En | MEDLINE | ID: mdl-37076317

Vasomotor symptoms (VMS) are characteristic of menopause experienced by over 75% of postmenopausal women with significant health and socioeconomic implications. Although the average duration of symptoms is seven years, 10% of women experience symptoms for more than a decade. Although menopausal hormone therapy (MHT) remains an efficacious and cost-effective treatment, its use may not be suitable in all women, such as those at an increased risk of breast cancer or gynaecological malignancy. The neurokinin B (NKB) signaling pathway, together with its intricate connection to the median preoptic nucleus (MnPO), has been postulated to provide integrated reproductive and thermoregulatory responses, with a central role in mediating postmenopausal VMS. This review describes the physiological hypothalamo-pituitary-ovary (HPO) axis, and subsequently the neuroendocrine changes that occur with menopause using evidence derived from animal and human studies. Finally, data from the latest clinical trials using novel therapeutic agents that antagonise NKB signaling are reviewed.


Hot Flashes , Menopause , Animals , Female , Humans , Hot Flashes/drug therapy , Hot Flashes/etiology , Hot Flashes/metabolism , Menopause/physiology , Neurokinin B/metabolism , Hormone Replacement Therapy , Signal Transduction
3.
Neuroendocrinology ; 112(7): 649-665, 2022.
Article En | MEDLINE | ID: mdl-34592740

INTRODUCTION: Menopausal hot flashes are related to hypothalamic preoptic area (POA) dysfunction. Thermosensitive transient receptor potential channels (ThermoTRPs) are involved in temperature sensing and regulation of thermosensitive neurons (TSNs) in the POA. Whether ThermoTRP-TSNs in the POA, particularly the non-noxious thermoreceptor, transient receptor potential melastatin 2 (TRPM2), are involved in the occurrence of hot flashes is still unclear. METHODS: Twenty wild-type and 50 Trpm2-Cre adult female mice were randomly divided into sham (SHAM) and ovariectomy (OVX) groups. In the POA, ERα, ERß, GPR30, TRPA1, TRPM8, TRPM2, and TRPV1 expression was detected by Western blot or/and quantitative real-time polymerase chain reaction and the number of TSNs expressing TRPM2 (TRPM2-TSNs) by immunofluorescence. Before and after TRPM2-TSN activation/inhibition, back (BST) and tail skin temperature (TST) and the proportion of glutamatergic and GABAergic neurons among TRPM2-TSNs were recorded. RESULTS: Compared with SHAM, the expression of ERα, ERß, TRPM2, and TRPM8 in the POA of the OVX group decreased, with a significantly larger change range for TRPM2 than TRPM8. In addition, the number of TRPM2-TSNs showing TRPA1, TRPM8, and TRPV1 expression in the OVX group decreased, and the proportion of glutamatergic and GABAergic neurons in TRPM2-TSNs decreased and increased, respectively. Meanwhile, BST and TST increased. After activating or inhibiting TRPM2-TSNs, the proportions of glutamatergic and GABAergic neurons in TRPM2-TSNs changed, along with the BST and TST. CONCLUSION: In menopause, the abnormal quantity and function of TRPM2-TSNs in the POA is key for the development of hot flashes, characterized by an imbalance in heat dissipation and production due to the corresponding imbalance in glutamatergic and GABAergic neurons.


TRPM Cation Channels , Animals , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Female , Hot Flashes/metabolism , Menopause , Mice , Neurons/metabolism , Preoptic Area/metabolism , TRPM Cation Channels/metabolism
5.
J Microbiol Biotechnol ; 30(2): 178-186, 2020 Feb 28.
Article En | MEDLINE | ID: mdl-31752065

Licorice (Glycyrrhiza uralensis) contains several compounds that have been reported to alleviate menopausal symptoms via interacting with estrogen receptors (ERs). The compounds exist mainly in the form of glycosides, which exhibit low bioavailability and function. To bioconvert liquiritin and isoliquiritin, the major estrogenic compounds, to the corresponding deglycosylated liquiritigenin and isoliquiritigenin, respectively, licorice was fermented with Monascus, which has been demonstrated to deglycosylate other substances. The contents of liquiritigenin and isoliquiritigenin in Monascus-fermented licorice increased by 10.46-fold (from 38.03 µM to 379.75 µM) and 12.50-fold (from 5.53 µM to 69.14 µM), respectively, compared with their contents in non-fermented licorice. Monascus-fermented licorice exhibited 82.5% of the ERß binding activity of that observed in the positive control (17 ß-estradiol), whereas the non-fermented licorice exhibited 54.1% of the binding activity in an in vivo ER binding assay. The increase in the ERß binding activity was associated with increases in liquiritigenin and isoliquiritigenin contents. Liquiritigenin acts as a selective ligand for ERß, which alleviates menopausal symptoms with fewer side effects, such as heart disease and hypertension, compared with a ligand for ERα. In addition, Monascus-fermented licorice contained 731 mg/kg of monacolin K, one of the metabolites produced by Monascus that reduces serum cholesterol. Therefore, Monascus-fermented licorice is a promising material for the prevention and treatment of menopausal syndrome with fewer side effects.


Biotransformation , Glycyrrhiza uralensis/chemistry , Glycyrrhiza/chemistry , Menopause/drug effects , Monascus/metabolism , Plant Extracts/pharmacology , Chalcones/chemistry , Chalcones/pharmacology , Chromatography, High Pressure Liquid , Drug Evaluation, Preclinical , Estrogen Receptor beta/metabolism , Fermentation , Flavanones/chemistry , Flavanones/pharmacology , Flavanones/therapeutic use , Glucosides/chemistry , Glucosides/pharmacology , Hot Flashes/drug therapy , Hot Flashes/metabolism , Lovastatin/chemistry , Lovastatin/pharmacology , Menopause/metabolism , Plant Extracts/chemistry , Plant Extracts/therapeutic use , Protein Binding
7.
Discov Med ; 27(149): 177-188, 2019 05.
Article En | MEDLINE | ID: mdl-31361980

Genistein is an isoflavone derived from soy-rich products, which is known to exhibit several beneficial biological effects, such as anti-tumor activity, improvement of glucose metabolism, and reduction of the frequency of peri-menopausal hot flashes, and thus has potential for clinical application. Certain limitations and side effects, such as low bioavailability, biological estrogenic activity, and detrimental effects on thyroid function, have restricted its clinical applications to some extent. Recently, it has been reported that fermentation, use of micromicelles, and modification of its chemical structure can enhance the bioavailability of genistein. Moreover, the modification of its molecular structure may also eliminate its biological estrogenic activity and adverse effects on thyroid function. In this review, we summarize the clinical application prospects and limitations of genistein, as well as the plausible solutions to overcome its low bioavailability, phytoestrogenic activity, and adverse effects on thyroid function.


Antineoplastic Agents, Phytogenic , Estrogens , Genistein , Hot Flashes/drug therapy , Menopause/metabolism , Thyroid Gland/metabolism , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/pharmacology , Estrogens/pharmacokinetics , Estrogens/therapeutic use , Female , Genistein/pharmacokinetics , Genistein/therapeutic use , Hot Flashes/metabolism , Hot Flashes/pathology , Humans , Micelles
8.
Endocrinology ; 160(4): 803-816, 2019 04 01.
Article En | MEDLINE | ID: mdl-30753503

We have proposed that arcuate neurons coexpressing kisspeptin, neurokinin B, and dynorphin (KNDy neurons) contribute to hot flushes via projections to neurokinin 3 receptor (NK3R)-expressing neurons in the median preoptic nucleus (MnPO). To characterize the thermoregulatory role of MnPO NK3R neurons in female mice, we ablated these neurons using injections of saporin toxin conjugated to a selective NK3R agonist. Loss of MnPO NK3R neurons increased the core temperature (TCORE) during the light phase, with the frequency distributions indicating a regulated shift in the balance point. The increase in TCORE in the ablated mice occurred despite changes in the ambient temperature and regardless of estrogen status. We next determined whether an acute increase in ambient temperature or higher TCORE would induce Fos in preoptic enhanced green fluorescent protein (EGFP)-immunoreactive neurons in Tacr3-EGFP mice. Fos activation was increased in the MnPO but no induction of Fos was found in NK3R (EGFP-immunoreactive) neurons. Thus, MnPO NK3R neurons are not activated by warm thermosensors in the skin or viscera and are not warm-sensitive neurons. Finally, RNAscope was used to determine whether Tacr3 (NK3R) mRNA was coexpressed with vesicular glutamate transporter 2 or vesicular γ-aminobutyric acid (GABA) transporter mRNA, markers of glutamatergic and GABAergic neurotransmission, respectively. In the MnPO, 94% of NK3R neurons were glutamatergic, but in the adjacent medial preoptic area, 97% of NK3R neurons were GABAergic. Thus, NK3R neurons in the MnPO are glutamatergic and play a role in reducing TCORE but are not activated by warm thermal stimuli (internal or external). These findings suggest that KNDy neurons modulate thermosensory pathways for heat defense indirectly via a subpopulation of glutamatergic MnPO neurons that express NK3R.


Body Temperature Regulation/physiology , Glutamic Acid/metabolism , Neurons/metabolism , Preoptic Area/metabolism , Receptors, Neurokinin-3/metabolism , Animals , Dynorphins/metabolism , Female , Hot Flashes/metabolism , Hot Temperature , Kisspeptins/metabolism , Mice , Neurokinin B/metabolism , Proto-Oncogene Proteins c-fos/metabolism
9.
Neuroendocrinology ; 109(3): 242-248, 2019.
Article En | MEDLINE | ID: mdl-30504731

Menopause is associated with significant symptomatic burden, with approximately two-thirds of postmenopausal women suffering from vasomotor symptoms, hot flushes, and night sweats. The mainstay of treatment for hot flushes continues to be hormone replacement therapy. However, as hormone replacement therapy is contraindicated in some cases, alternative, efficacious treatment options are also required. Hot flushes are thought to arise as a result of significant changes in the neuroendocrine circuitry underpinning the reproductive axis during menopause. This includes reduced circulating ovarian oestrogens, hypersecretion of gonadotropins, and increased expression of kisspeptin and neurokinin B (NKB) within the infundibular nucleus of the hypothalamus. In recent years, NKB, predominantly acting via the neurokinin 3 receptor (NK3R), has emerged as an important player in the development of menopausal hot flushes. Antagonism of NK3R has garnered much interest as a novel therapeutic target to help ameliorate hot flush symptoms. Improvements in hot flush frequency, severity, and quality of life have been demonstrated in a number of clinical trials using novel NK3R antagonists in postmenopausal women. Within this review, we will explore the growing body of evidence supporting antagonism of NK3R as a potentially promising treatment for menopausal hot flushes.


Hot Flashes/drug therapy , Menopause/drug effects , Receptors, Neurokinin-3/antagonists & inhibitors , Animals , Female , Hot Flashes/etiology , Hot Flashes/metabolism , Humans , Menopause/metabolism , Receptors, Neurokinin-3/metabolism
11.
Neuroendocrinology ; 106(2): 148-157, 2018.
Article En | MEDLINE | ID: mdl-28380486

OBJECTIVES: Neurokinin B (NKB) and kisspeptin are obligate for normal gonadotropin secretion, and links between gonadotropin-releasing hormone (GnRH) pulsatility and vasomotor symptoms have been proposed. Using a selective NKB receptor (NK3R) antagonist, the role of NKB in the hypergonadotropic state in menopausal women was explored. METHODS: Eleven postmenopausal women were administered the NK3R antagonist MLE4901 at 40 mg twice daily orally for 7 days. Ten-minute blood sampling for 8 h was performed before and on the last day of NK3R antagonist treatment for luteinising hormone (LH) pulsatility analysis with kisspeptin-10 (0.3 µg/kg i.v. bolus) administered at 6 h on both days. Hot flash frequency and severity were self-reported for 7 days before and during NK3R antagonist administration. RESULTS: LH fell from 29.3 ± 4.1 to 24.4 ± 3.8 IU/L (p < 0.05) after 7 days of NK3R antagonist treatment, with no change in follicle-stimulating hormone (FSH). Basal (non-pulsatile) LH secretion was reduced (549.0 ± 70.8 vs. 366.1 ± 92.1 IU/L/6 h, p = 0.006), and while the LH pulse frequency did not change in the group as a whole (from 0.8 ± 0.1 to 0.7 ± 0.1 pulses/h, ns), it did fall in the 8 women with hot flashes (from 1.0 ± 0.1 to 0.7 ± 0.1 pulses/h, p < 0.05). These women also reported a reduction in hot flash frequency (from 3.4 ± 1.2 to 1.0 ± 0.6 hot flashes/day, p = 0.008) whilst taking the NK3R antagonist. Kisspeptin-10 did not affect LH secretion with or without the NK3R antagonist. CONCLUSIONS: The administration of an NK3R antagonist indicates a role for NKB in the regulation of LH/GnRH in postmenopausal women, whereas the lack of response to kisspeptin may reflect the hypo-oestrogenic state. These data support a link of LH/GnRH pulsatility and vasomotor symptoms with NK3R antagonism as a potential therapeutic approach.


Gonadotropins/metabolism , Hot Flashes/drug therapy , Neurokinin B/metabolism , Postmenopause/drug effects , Receptors, Neurokinin-3/antagonists & inhibitors , Administration, Oral , Female , Follicle Stimulating Hormone/blood , Gonadotropin-Releasing Hormone/metabolism , Hot Flashes/metabolism , Humans , Kisspeptins/metabolism , Kisspeptins/therapeutic use , Luteinizing Hormone/blood , Middle Aged , Periodicity , Postmenopause/metabolism , Receptors, Neurokinin-3/metabolism , Treatment Outcome
12.
Hum Reprod Update ; 23(5): 600-628, 2017 09 01.
Article En | MEDLINE | ID: mdl-28903474

BACKGROUND: Although some unequivocal underlying mechanisms of menopausal hot flushes have been demonstrated in animal models, the paucity of similar approaches in humans impedes further mechanistic outcomes. Human studies might show some as yet unexpected physiological mechanisms of metabolic adaptation that permeate the phase of decreased oestrogen levels in both symptomatic and asymptomatic women. This is particularly relevant because both the severity and time span of hot flushes are associated with increased risk of chronic inflammatory disease. On the other hand, oestrogen induces the expression of heat shock proteins of the 70 kDa family (HSP70), which are anti-inflammatory and cytoprotective protein chaperones, whose expression is modulated by different types of physiologically stressful situations, including heat stress and exercise. Therefore, lower HSP70 expression secondary to oestrogen deficiency increases cardiovascular risk and predisposes the patient to senescence-associated secretory phenotype (SASP) that culminates in chronic inflammatory diseases, such as obesities, type 2 diabetes, neuromuscular and neurodegenerative diseases. OBJECTIVE AND RATIONALE: This review focuses on HSP70 and its accompanying heat shock response (HSR), which is an anti-inflammatory and antisenescent pathway whose intracellular triggering is also oestrogen-dependent via nitric oxide (NO) production. The main goal of the manuscript was to show that the vasomotor symptoms that accompany hot flushes may be a disguised clue for important neuroendocrine alterations linking oestrogen deficiency to the anti-inflammatory HSR. SEARCH METHODS: Results from our own group and recent evidence on hypothalamic control of central temperature guided a search on PubMed and Google Scholar websites. OUTCOMES: Oestrogen elicits rapid production of the vasodilatory gas NO, a powerful activator of HSP70 expression. Whence, part of the protective effects of oestrogen over cardiovascular and neuroendocrine systems is tied to its capacity of inducing the NO-elicited HSR. The hypothalamic areas involved in thermoregulation (infundibular nucleus in humans and arcuate nucleus in other mammals) and whose neurons are known to have their function altered after long-term oestrogen ablation, particularly kisspeptin-neurokinin B-dynorphin neurons, (KNDy) are the same that drive neuroprotective expression of HSP70 and, in many cases, this response is via NO even in the absence of oestrogen. From thence, it is not illogical that hot flushes might be related to an evolutionary adaptation to re-equip the NO-HSP70 axis during the downfall of circulating oestrogen. WIDER IMPLICATIONS: Understanding of HSR could shed light on yet uncovered mechanisms of menopause-associated diseases as well as on possible manipulation of HSR in menopausal women through physiological, pharmacological, nutraceutical and prebiotic interventions. Moreover, decreased HSR indices (that can be clinically determined with ease) in perimenopause could be of prognostic value in predicting the moment and appropriateness of starting a HRT.


Heat-Shock Response , Hot Flashes/metabolism , Menopause/physiology , Nitric Oxide/metabolism , Animals , Estrogens/physiology , Female , Heat-Shock Proteins/metabolism , Humans , Hypothalamus , Nitric Oxide/blood , Risk Factors
13.
PLoS One ; 12(4): e0176430, 2017.
Article En | MEDLINE | ID: mdl-28448547

INTRODUCTION: Hot flashes have been postulated to be linked to the development of metabolic disorders. This study aimed to evaluate the relationship between hot flashes, adipocyte-derived hormones, and insulin resistance in healthy, non-obese postmenopausal women. PARTICIPANTS AND DESIGN: In this cross-sectional study, a total of 151 women aged 45-60 years were stratified into one of three groups according to hot-flash status over the past three months: never experienced hot flashes (Group N), mild-to-moderate hot flashes (Group M), and severe hot flashes (Group S). Variables measured in this study included clinical parameters, hot flash experience, fasting levels of circulating glucose, lipid profiles, plasma insulin, and adipocyte-derived hormones. Multiple linear regression analysis was used to evaluate the associations of hot flashes with adipocyte-derived hormones, and with insulin resistance. SETTINGS: The study was performed in a hospital medical center. RESULTS: The mean (standard deviation) of body-mass index was 22.8(2.7) for Group N, 22.6(2.6) for Group M, and 23.5(2.4) for Group S, respectively. Women in Group S displayed statistically significantly higher levels of leptin, fasting glucose, and insulin, and lower levels of adiponectin than those in Groups M and N. Multivariate linear regression analysis revealed that hot-flash severity was significantly associated with higher leptin levels, lower adiponectin levels, and higher leptin-to-adiponectin ratio. Univariate linear regression analysis revealed that hot-flash severity was strongly associated with a higher HOMA-IR index (% difference, 58.03%; 95% confidence interval, 31.00-90.64; p < 0.001). The association between hot flashes and HOMA-IR index was attenuated after adjusting for leptin or adiponectin and was no longer significant after simultaneously adjusting for leptin and adiponectin. CONCLUSION: The present study provides evidence that hot flashes are associated with insulin resistance in postmenopausal women. It further suggests that hot flash association with insulin resistance is dependent on the combination of leptin and adiponectin variables.


Adiponectin/blood , Hot Flashes/blood , Hot Flashes/metabolism , Insulin Resistance , Leptin/blood , Postmenopause/blood , Postmenopause/metabolism , Adipocytes/metabolism , Female , Hot Flashes/pathology , Humans , Middle Aged
14.
Maturitas ; 96: 26-32, 2017 Feb.
Article En | MEDLINE | ID: mdl-28041591

OBJECTIVES: This study examined the relationship between salivary cortisol levels and hot flashes during midlife. Previous studies have shown that cortisol levels increase with hot flashes in the laboratory, and higher cortisol levels have been associated with more severe hot flashes. Salivary cortisol levels were also examined in relation to total number of midlife symptoms. METHODS: Women aged 40-60 years (n=109) reported the presence or absence of 23 symptoms, including hot flashes, during the previous 2 weeks. Salivary samples were collected at waking, 30min after waking, 1h before bedtime, and at bedtime. The cortisol awakening response (CAR), cortisol daily decline (CDD), log transformed salivary cortisol levels at each time point, and mean cortisol levels were compared by hot flash report using t-tests. Logistic regression analyses were performed to assess the association between each cortisol measure and the presence or absence of hot flashes, after controlling for potential covariates. RESULTS: Salivary cortisol levels were not significantly associated with hot flashes or sum of symptoms. Hot flash report did not differentiate women who had a positive CAR from those who did not, or women who showed strong CDD from those who did not. CONCLUSION: Symptomatic women - defined by hot flash report or symptom total - were not found to have higher salivary cortisol levels.


Hot Flashes/metabolism , Hydrocortisone/metabolism , Menopause/metabolism , Saliva/metabolism , Adult , Female , Hot Flashes/complications , Humans , Middle Aged , Time Factors
16.
J Clin Endocrinol Metab ; 101(10): 3847-3855, 2016 10.
Article En | MEDLINE | ID: mdl-27680875

CONTEXT: Women are at increased risk for mood disturbance during the menopause transition. Hot flashes (HFs), sleep disruption, and fluctuating estradiol levels correlate with menopause-associated depression but co-occur, making cause and effect relationships difficult to disentangle. OBJECTIVE: Using a GnRH agonist (GnRHa) experimental model, we investigated whether depressive symptoms are associated with HFs and/or are explained by concomitant sleep fragmentation in the absence of estradiol fluctuation. DESIGN AND INTERVENTION: Depressive symptoms, objective polysomnographic sleep parameters, subjective sleep quality, serum estradiol, and HFs were assessed before and 4 weeks after open-label depot GnRHa (leuprolide 3.75-mg) administration. SETTING: Academic medical center. PARTICIPANTS: Twenty-nine healthy nondepressed premenopausal volunteers (mean age, 27.3 years). RESULTS: Serum estradiol was rapidly and uniformly suppressed. HFs developed in 69% of the subjects. On univariate analysis, worsening of mood was predicted by increases in time in light sleep (stage N1), number of transitions to wake, non-REM arousals, subjective sleep quality, and reductions in perceived sleep efficiency (all P < .045), as well as the number of nighttime (P = .006), but not daytime (P = .28), HFs reported. In adjusted models, the number of nighttime HFs reported, increases in non-REM arousals, time in stage N1, transitions to wake, and reduced sleep quality remained significant predictors of mood deterioration (P ≤ .05). CONCLUSIONS: Depressive symptoms emerged after estradiol withdrawal in association with objectively and subjectively measured sleep disturbance and the number of nighttime, but not daytime, HFs reported. Results suggest that sleep disruption and perceived nighttime HFs both contribute to vulnerability to menopause-associated depressive symptoms in hypoestrogenic women.


Depression/metabolism , Estradiol/metabolism , Gonadotropin-Releasing Hormone/agonists , Hot Flashes/metabolism , Sleep Wake Disorders/metabolism , Adolescent , Adult , Depression/chemically induced , Estradiol/blood , Fertility Agents, Female/administration & dosage , Fertility Agents, Female/pharmacology , Gonadotropin-Releasing Hormone/pharmacology , Healthy Volunteers , Hot Flashes/chemically induced , Humans , Leuprolide/administration & dosage , Leuprolide/pharmacology , Menopause/metabolism , Middle Aged , Sleep Wake Disorders/chemically induced , Young Adult
17.
Ann Anat ; 208: 31-39, 2016 Nov.
Article En | MEDLINE | ID: mdl-27562857

Perimenopausal syndromes begin as ovarian function ceases and the most common symptoms are hot flushes. Data indicate that the projections of serotonin to hypothalamus may be involved in the mechanism of hot flushes. Therefore, the aim of this study is to investigate the potential role of the serotonin dorsal raphe-preoptic hypothalamus pathway for hot flushes in an animal model of menopause. We determined the changes in serotonin expression in the dorsal raphe (DR) and preoptic anterior hypothalamus (POAH) in ovariectomized rats. We also explored the therapeutical effects of estradiol valerate and Remifemin in this model. Eighty female Sprague-Dawley rats were randomly assigned to sham-operated (SHAM) group, ovariectomy (OVX) group with vehicle, ovariectomy with estradiol valerate treatment (OVX+E) group and ovariectomy with Remifemin (OVX+ICR) group. Serotonin expression was evaluated in the DR and POAH using immunofluorescence and quantified in the DR using an enzyme-linked immunosorbent assay (ELISA). Apoptosis was analyzed in the DR by TUNEL assay. The number of serotonin immunoreactive neurons and the level of serotonin expression in the DR decreased significantly following OVX compared to the SHAM group. No TUNEL-positive cells were detected in the DR in any group. In addition, following OVX, the number of serotonin-positive fibers decreased significantly in the ventromedial preoptic nucleus (VMPO), especially in the ventrolateral preoptic nucleus (VLPO). Treatment with either estradiol or Remifemin for 4 weeks countered the OVX-induced decreases in serotonin levels in both the DR and the hypothalamus, with levels in the treated rats similar to those in the SHAM group. A fluorescently labeled retrograde tracer was injected into the VLPO at the 4-week time point. A significantly lower percentage of serotonin with CTB double-labeled neurons in CTB-labeled neurons was demonstrated after ovariectomy, and both estradiol and Remifemin countered this OVX-induced decrease. We conclude that serotonin pathway is changed after ovariectomy, including the serotonin synthesis in DR and serotonin fibers in PO/AH, both E and Remifemin have an equivalent therapeutic effect on it.


Dorsal Raphe Nucleus/metabolism , Estradiol/analogs & derivatives , Hot Flashes/metabolism , Hypothalamus/metabolism , Plant Extracts/administration & dosage , Preoptic Area/metabolism , Serotonin/metabolism , Animals , Cimicifuga , Dorsal Raphe Nucleus/cytology , Dorsal Raphe Nucleus/drug effects , Down-Regulation/drug effects , Estradiol/administration & dosage , Female , Hot Flashes/drug therapy , Hypothalamus/cytology , Hypothalamus/drug effects , Menopause/drug effects , Menopause/metabolism , Neural Pathways/cytology , Neural Pathways/metabolism , Ovariectomy , Preoptic Area/cytology , Preoptic Area/drug effects , Rats , Rats, Sprague-Dawley , Serotonergic Neurons/cytology , Serotonergic Neurons/metabolism , Tissue Distribution
18.
J Med Food ; 19(3): 228-37, 2016 Mar.
Article En | MEDLINE | ID: mdl-26848802

Many natural substances were screened to develop nutraceuticals that reduce menopausal symptoms. A complex of Cirsium japonicum var. maackii and Thymus vulgaris extracts, named MS-10, had significant positive effects. Under a low concentration of estrogen, which represents postmenopausal physiological conditions, MS-10 had beneficial effects on estrogen receptor-expressing MCF-7 cells by reversibly enhancing estrogen activity. In addition, in the ovariectomized rat model, changes in bone-specific alkaline phosphatase activity and osteocalcin, as well as low-density lipoprotein cholesterol and triglyceride levels were significantly decreased by MS-10. These results show that MS-10 protected bone health and reduced metabolic disturbances. Furthermore, in a clinical study, all menopausal symptoms, including hot flushes, parenthesis, insomnia, nervousness, melancholia, vertigo, fatigue, rheumatic pain, palpitations, formication, and headache, as well as colpoxerosis, were significantly improved by taking MS-10 for 90 days. Therefore, the evidence supports that MS-10 is an effective natural substance that can safely improve menopausal symptoms, including colpoxerosis.


Cirsium/chemistry , Menopause/drug effects , Plant Extracts/administration & dosage , Thymus Plant/chemistry , Vaginal Diseases/prevention & control , Animals , Female , Hot Flashes/drug therapy , Hot Flashes/metabolism , Hot Flashes/prevention & control , Humans , Lipoproteins, LDL/metabolism , Menopause/metabolism , Middle Aged , Osteocalcin/metabolism , Rats , Rats, Sprague-Dawley , Vaginal Diseases/drug therapy , Vaginal Diseases/metabolism
19.
Psychoneuroendocrinology ; 66: 1-10, 2016 Apr.
Article En | MEDLINE | ID: mdl-26766119

Hot flashes, hormones, and psychosocial factors contribute to insomnia risk in the context of the menopausal transition. Stress is a well-recognized factor implicated in the pathophysiology of insomnia; however the impact of stress on sleep and sleep-related processes in perimenopausal women remains largely unknown. We investigated the effect of an acute experimental stress (impending Trier Social Stress Task in the morning) on pre-sleep measures of cortisol and autonomic arousal in perimenopausal women with and without insomnia that developed in the context of the menopausal transition. In addition, we assessed the macro- and micro-structure of sleep and autonomic functioning during sleep. Following adaptation to the laboratory, twenty two women with (age: 50.4 ± 3.2 years) and eighteen women without (age: 48.5 ± 2.3 years) insomnia had two randomized in-lab overnight recordings: baseline and stress nights. Anticipation of the task resulted in higher pre-sleep salivary cortisol levels and perceived tension, faster heart rate and lower vagal activity, based on heart rate variability measures, in both groups of women. The effect of the stress manipulation on the autonomic nervous system extended into the first 4 h of the night in both groups. However, vagal tone recovered 4-6 h into the stress night in controls but not in the insomnia group. Sleep macrostructure was largely unaltered by the stress, apart from a delayed latency to REM sleep in both groups. Quantitative analysis of non-rapid eye movement sleep microstructure revealed greater electroencephalographic (EEG) power in the beta1 range (15-≤23 Hz), reflecting greater EEG arousal during sleep, on the stress night compared to baseline, in the insomnia group. Hot flash frequency remained similar on both nights for both groups. These results show that pre-sleep stress impacts autonomic nervous system functioning before and during sleep in perimenopausal women with and without insomnia. Findings also indicate that women with insomnia had increased EEG arousal and lacked recovery in vagal activity across the stress night suggesting a greater sensitivity to stress in this group.


Autonomic Nervous System/physiopathology , Menopause/physiology , Sleep/physiology , Stress, Psychological/physiopathology , Acute Disease , Autonomic Nervous System/metabolism , Electroencephalography , Female , Heart Rate/physiology , Hot Flashes/metabolism , Hot Flashes/physiopathology , Humans , Hydrocortisone/metabolism , Middle Aged , Polysomnography , Sleep Initiation and Maintenance Disorders/metabolism , Sleep Initiation and Maintenance Disorders/physiopathology , Stress, Psychological/metabolism , Vagus Nerve/physiology
20.
Clin Chem Lab Med ; 54(5): 739-53, 2016 May.
Article En | MEDLINE | ID: mdl-26544103

The postmenopausal phase of life is frequently associated in women with subjective symptoms (e.g. vasomotor) and real diseases (atherosclerosis with coronary ischemia, osteoporosis, Alzheimer-type neurodegeneration, urogenital dystrophy), which together determine the post-menopausal syndrome. Observations that oxidative damage by reactive oxygen/nitrogen species in experimental models can contribute to the pathogenesis of these disturbances stimulated research on the relationships between menopause, its endocrine deficiency, oxidative balance and the "wellness" in postmenopausal life. The connection among these events is probably due to the loss of protective actions exerted by estrogens during the fertile life. Most recent studies have revealed that estrogens exert an antioxidant action not by direct chemical neutralization of reactants as it was expected until recently but by modulating the expression of antioxidant enzymes that control levels of biological reducing agents. Also nutritional antioxidants apparently act by a similar mechanism. From this perspective it is conceivable that a cumulative control of body oxidant challenges and biological defenses could help in monitoring between "normal" and "pathological" menopause. However, as clinical studies failed to confirm this scenario in vivo, we have decided to review the existing literature to understand the causes of this discrepancy and whether this was due to methodologic reasons or to real failure of the basic hypothesis.


Atherosclerosis/metabolism , Coronary Artery Disease/metabolism , Hot Flashes/metabolism , Menopause/metabolism , Neurodegenerative Diseases/metabolism , Osteoporosis/metabolism , Oxidative Stress , Female , Humans
...