Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
J Ethnopharmacol ; 290: 115047, 2022 May 23.
Article in English | MEDLINE | ID: mdl-35122976

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Guilingji (GLJ), which has been used to treat male diseases in China for centuries, contains 28 Chinese herbs and was previously established as an effective treatment for male sexual dysfunction. However, its mechanism of action remains unclear. AIM OF THE STUDY: To explore the efficacy and mechanism of action of GLJ in improving senile sexual dysfunction (SSD) in aging rats. MATERIALS AND METHODS: An aging rat model of SSD was induced by the subcutaneous injection of d-galactose (300 mg⋅kg-1) and used to analyse the effects of GLJ (different concentrations of 37.5, 75, and 150 mg⋅kg-1) on the mating of aging rats. At the end of the 8th week, histopathological analysis of testicular tissues, assessment of the hypothalamic-pituitary-gonadal (HPG) axis hormone levels in serum or brain, and metabonomics analysis of the brain and testicular tissue with liquid chromatography-mass spectrometry was performed to explore the mechanism of action of GLJ. RESULT: After treatment with GLJ, the mount and ejaculation latency levels were increased in the treatment group than those in model group (P < 0.05), moreover, the testicular morphology was improved. Gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH) levels in rats were also improved significant (P < 0.05) compared with those in the model group. Furthermore, the metabonomics results in the testicular and brain tissue showed that GLJ improved SSD by adjusting amino acid and lipid metabolism. CONCLUSION: This study integrated the complementary metabolic profiles of the target tissues. GLJ might affect SSD rats by regulating amino acid and lipid metabolism and may modulate sensitivity to the signaling pathway in the HPG axis. This study provides an essential basis for the broad clinical application of GLJ.


Subject(s)
Aging/pathology , Brain/drug effects , Drugs, Chinese Herbal/pharmacology , Sexual Behavior, Animal/drug effects , Sexual Dysfunction, Physiological/pathology , Testis/drug effects , Amino Acids/drug effects , Amino Acids/metabolism , Animals , Disease Models, Animal , Female , Gonadotropin-Releasing Hormone/drug effects , Lipid Metabolism/drug effects , Luteinizing Hormone/drug effects , Male , Metabolomics , Rats , Rats, Sprague-Dawley
2.
Reprod Biol Endocrinol ; 19(1): 120, 2021 Aug 03.
Article in English | MEDLINE | ID: mdl-34344365

ABSTRACT

BACKGROUND: This study aimed to detect the effect of angiotensin receptor 1 (AT1) knock out (KO) on spermatogenesis and hypothalamic-pituitary-gonadal (HPG) axis hormone expression. METHODS: Normal C57BL/6 male mice were used as control group or treated with angiotensin receptor blocker, in addition heterozygous ± AT1KO mice were generated. After caged at a ratio of 2 to 1 with females, pregnancy rates of female mice were determined by detection of vaginal plugs. Deformity rate of spermatozoa was evaluated by eosin staining and morphology evaluation. The AT1 mRNA expression in the testes of male ± AT1KO mice was detected by quantitative real-time polymerase chain reaction (QRT-PCR). Serum GnRH level was determined by ELISA. RESULTS: Compared to control, ± AT1KO mice showed reduced expression of AT1 in testes, pituitary and hypothalamus. In addition, decreased level of GnRH, but not follicle stimulating hormone (FSH) or luteinizing hormone (LH), in ± AT1KO mice was detected. Treatment with angiotensin receptor blocker (ARB) did not have significant effects on HPG hormones. ± AT1KO mice exhibited male infertility and significant abnormality of sperm morphology. CONCLUSION: Reduced AT1 knockout resulted in male infertility, potentially by inducing abnormal spermatogenesis. Both testis and HPG axis signaling may be involved.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Infertility, Male/genetics , Receptor, Angiotensin, Type 1/genetics , Spermatogenesis/genetics , Testis/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Gonadotropin-Releasing Hormone/drug effects , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Infertility, Male/metabolism , Losartan/pharmacology , Male , Mice , Mice, Knockout , Pituitary Gland/drug effects , Pituitary Gland/metabolism , Receptor, Angiotensin, Type 1/metabolism , Spermatogenesis/drug effects , Testis/drug effects
3.
Nat Rev Endocrinol ; 17(2): 83-96, 2021 02.
Article in English | MEDLINE | ID: mdl-33288917

ABSTRACT

The onset of puberty and the female ovulatory cycle are important developmental milestones of the reproductive system. These processes are controlled by a tightly organized network of neurotransmitters and neuropeptides, as well as genetic, epigenetic and hormonal factors, which ultimately drive the pulsatile secretion of gonadotropin-releasing hormone. They also strongly depend on organizational processes that take place during fetal and early postnatal life. Therefore, exposure to environmental pollutants such as endocrine-disrupting chemicals (EDCs) during critical periods of development can result in altered brain development, delayed or advanced puberty and long-term reproductive consequences, such as impaired fertility. The gonads and peripheral organs are targets of EDCs, and research from the past few years suggests that the organization of the neuroendocrine control of reproduction is also sensitive to environmental cues and disruption. Among other mechanisms, EDCs interfere with the action of steroidal and non-steroidal receptors, and alter enzymatic, metabolic and epigenetic pathways during development. In this Review, we discuss the cellular and molecular consequences of perinatal exposure (mostly in rodents) to representative EDCs with a focus on the neuroendocrine control of reproduction, pubertal timing and the female ovulatory cycle.


Subject(s)
Endocrine Disruptors/pharmacology , Environmental Exposure , Epigenesis, Genetic/drug effects , Estradiol/metabolism , Gonadotropin-Releasing Hormone/drug effects , Hypothalamus/drug effects , Neurons/drug effects , Animals , Cell Movement , DNA Methylation/drug effects , Feedback, Physiological/drug effects , Female , GABA Agents/metabolism , Germ Cells/metabolism , Glutamic Acid/metabolism , Gonadotropin-Releasing Hormone/metabolism , Histone Code/drug effects , Humans , Hypothalamus/cytology , Hypothalamus/growth & development , Hypothalamus/metabolism , Kisspeptins/metabolism , Male , Neurons/metabolism , Ovulation/drug effects , Ovulation/metabolism , Pregnancy , Prenatal Exposure Delayed Effects
4.
Genes (Basel) ; 11(7)2020 07 07.
Article in English | MEDLINE | ID: mdl-32645906

ABSTRACT

Semen changes the gene expression in endometrial and oviductal tissues modulating important processes for reproduction. We tested the hypothesis that mating and/or sperm-free seminal plasma deposition in the reproductive tract affect the expression of genes associated with sperm-lining epithelium interactions, ovulation, and pre-implantation effects (nerve growth factor, NGF; α/ß hydrolase domain-containing protein 2, ABHD2; C-terminal tensin-like protein, CTEN or TNS4; and versican, VCAN) in the period 10-72 h post-mating. In Experiment 1, does (n = 9) were treated with gonadotropin-releasing hormone (GnRH) (control), GnRH-stimulated, and vaginally infused with sperm-free seminal plasma (SP-AI), or GnRH-stimulated and naturally mated (NM). In Experiment 2, does (n = 15) were GnRH-stimulated and naturally mated. Samples were retrieved from the internal reproductive tracts (cervix-to-infundibulum) 20 h post-treatment (Experiment 1) or sequentially collected at 10, 24, 36, 68, or 72 h post-mating (Experiment 2, 3 does/period). All samples were processed for gene expression analysis by quantitative PCR. Data showed an upregulation of endometrial CTEN and NGF by NM, but not by SP-AI. The findings suggest that the NGF gene affects the reproductive tract of the doe during ovulation and beyond, influencing the maternal environment during early embryonic development.


Subject(s)
Endometrium/metabolism , Nerve Growth Factor/genetics , Serine Proteases/genetics , Sperm-Ovum Interactions , Tensins/genetics , Versicans/genetics , Animals , Endometrium/drug effects , Female , Gonadotropin-Releasing Hormone/drug effects , Male , Nerve Growth Factor/metabolism , Rabbits , Semen/metabolism , Serine Proteases/metabolism , Tensins/metabolism , Versicans/metabolism
5.
Am J Physiol Endocrinol Metab ; 319(1): E81-E90, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32396496

ABSTRACT

We have previously shown that systemic injection of erythropoietin-producing hepatocellular receptor A7 (EPHA7)-Fc raises serum luteinizing hormone (LH) levels before ovulation in female rats, indicating the induction of EPHA7 in ovulation. In this study, we aimed to identify the mechanism and hypothalamus-pituitary-ovary (HPO) axis level underlying the promotion of LH secretion by EPHA7. Using an ovariectomized (OVX) rat model, in conjunction with low-dose 17ß-estradiol (E2) treatment, we investigated the association between EPHA7-ephrin (EFN)A5 signaling and E2 negative feedback. Various rat models (OVX, E2-treated OVX, and abarelix treated) were injected with the recombinant EPHA7-Fc protein through the caudal vein to investigate the molecular mechanism underlying the promotion of LH secretion by EPHA7. Efna5 was observed strongly expressed in the arcuate nucleus of the female rat by using RNAscope in situ hybridization. Our results indicated that E2, combined with estrogen receptor (ER)α, but not ERß, inhibited Efna5 and gonadotropin-releasing hormone 1 (Gnrh1) expressions in the hypothalamus. In addition, the systemic administration of EPHA7-Fc restrained the inhibition of Efna5 and Gnrh1 by E2, resulting in increased Efna5 and Gnrh1 expressions in the hypothalamus as well as increased serum LH levels. Collectively, our findings demonstrated the involvement of EPHA7-EFNA5 signaling in the regulation of LH and the E2 negative feedback pathway in the hypothalamus, highlighting the functional role of EPHA7 in female reproduction.


Subject(s)
Ephrin-A5/metabolism , Estrogen Receptor alpha/metabolism , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Luteinizing Hormone/metabolism , Protein Precursors/metabolism , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Ephrin-A5/drug effects , Ephrin-A5/genetics , Estradiol/pharmacology , Estrogen Receptor beta/metabolism , Estrogens/pharmacology , Feedback, Physiological/drug effects , Feedback, Physiological/physiology , Female , Gonadotropin-Releasing Hormone/drug effects , Hormone Antagonists/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Hypothalamus/drug effects , Luteinizing Hormone/drug effects , Oligopeptides/pharmacology , Ovariectomy , Ovary/drug effects , Ovary/metabolism , Protein Precursors/drug effects , Rats , Receptor, EphA7/genetics , Receptor, EphA7/metabolism , Receptor, EphA7/pharmacology , Recombinant Proteins
6.
Med Sci Monit ; 26: e922860, 2020 May 26.
Article in English | MEDLINE | ID: mdl-32451371

ABSTRACT

BACKGROUND This study was designed to investigate the effect of high-glucose and high-fat condition on estrogen receptor- and sexual precocity-related genes in GT1-7 cells. MATERIAL AND METHODS In this study, CCK8 was used to detect cell viability, and TUNEL assay was used to detect apoptosis levels of GT1-7 cells after treatment with glucosamine and palmitate. The expression level of GnRH was measured by ELISA and RT-qPCR. RT-qPCR and Western blot were used to detect the expression of ERß, CD36, and GPR54 in GT1-7 cells, and the expression of ERß was detected using immunohistochemistry analysis. Finally, after adding the intervening drug tamoxifen to GT1-7 cells, the expression level of GnRH was measured by ELISA and Western blot analysis was used to detect the expression of GPR54 and GnRH. RESULTS GnRH secretion in the high-fat and high-glucose group increased continuously over time and peaked at 18 h, and GnRH gene expression peaked at 12 h. High-fat and high-glucose conditions also significantly increased the levels of estrogen receptors ß (ERß), fatty acid translocase protein (CD36), and G Protein-Coupled Receptors 54 (GPR54) in GT1-7 cells. After estrogen receptors ß (ER) was inhibited, GnRH secretion and GPR54 expression were decreased at 12 h and 18 h. CONCLUSIONS Our study demonstrates that high-glucose and high-fat conditions promote the secretion of GnRH and ER and the expression of genes related to sexual precocity in GT1-7 cells.


Subject(s)
Gene Expression/drug effects , Puberty, Precocious/genetics , Receptors, Estrogen/genetics , Animals , Apoptosis/drug effects , CD36 Antigens/genetics , Cell Line , Cell Survival/drug effects , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Estrogens/metabolism , Gene Expression/genetics , Glucosamine/pharmacology , Glucose/metabolism , Gonadotropin-Releasing Hormone/drug effects , Gonadotropin-Releasing Hormone/genetics , Kisspeptins/genetics , Mice , Palmitates/pharmacology , Puberty, Precocious/metabolism , Receptors, Estrogen/metabolism , Receptors, Kisspeptin-1/genetics
7.
Aging (Albany NY) ; 12(3): 2101-2122, 2020 02 02.
Article in English | MEDLINE | ID: mdl-32007953

ABSTRACT

As the elderly population grows, chronic metabolic dysfunction including obesity and diabetes are becoming increasingly common comorbidities. Hypothalamic inflammation through CNS resident microglia serves as a common pathway between developing obesity and developing systemic aging pathologies. Despite understanding aging as a life-long process involving interactions between individuals and their environment, limited studies address the dynamics of environment interactions with aging or aging therapeutics. We previously demonstrated environmental enrichment (EE) is an effective model for studying improved metabolic health and overall healthspan in mice, which acts through a brain-fat axis. Here we investigated the CSF1R inhibitor PLX5622 (PLX), which depletes microglia, and its effects on metabolic decline in aging in interaction with EE. PLX in combination with EE substantially improved metabolic outcomes in middle-aged female mice over PLX or EE alone. Chronic PLX treatment depleted 75% of microglia from the hypothalamus and reduced markers of inflammation without affecting brain-derived neurotrophic factor levels induced by EE. Adipose tissue remodeling and adipose tissue macrophage modulation were observed in response to CSF1R inhibition, which may contribute to the combined benefits seen in EE with PLX. Our study suggests benefits exist from combined drug and lifestyle interventions in aged animals.


Subject(s)
Adipose Tissue/drug effects , Aging/metabolism , Housing, Animal , Microglia/drug effects , Organic Chemicals/pharmacology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Social Environment , Adipose Tissue/metabolism , Animals , Body Composition/drug effects , Body Weight/drug effects , Brain-Derived Neurotrophic Factor/drug effects , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Corticotropin-Releasing Hormone/drug effects , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Female , Glial Fibrillary Acidic Protein/drug effects , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Glucose Tolerance Test , Gonadotropin-Releasing Hormone/drug effects , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Inflammation/genetics , Inflammation/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Neuropeptide Y/drug effects , Neuropeptide Y/genetics , Pro-Opiomelanocortin/drug effects , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Protein Kinase Inhibitors/pharmacology , Transcriptome/drug effects , Weight Loss
8.
Clin Pharmacokinet ; 59(3): 297-309, 2020 03.
Article in English | MEDLINE | ID: mdl-31749075

ABSTRACT

The clinical pharmacology of elagolix was extensively evaluated in clinical studies in healthy subjects and in women with endometriosis. Elagolix pharmacokinetics (PK) show significant population variability, however they are minimally affected by patients' baseline characteristics and demographics, except for clinically relevant extrinsic and intrinsic factors such as coadministrated strong organic anion transporting polypeptide (OATP) 1B1 inhibitors and severe hepatic impairment, which are contraindications for the use of elagolix. These studies enabled a comprehensive understanding of elagolix mechanism of action and the downstream pharmacodynamic (PD) effects on gonadotropin and ovarian hormones, as well as full characterization of the PK/PD (PKPD) relationships of elagolix at various dosages, including the approved 150 mg once daily and 200 mg twice daily dosing regimens for the management of moderate to severe pain associated with endometriosis. Several model-based analyses have contributed to understanding of the benefit-risk profile of elagolix in patients with endometriosis, through characterization of the exposure relationship with responder rates, with changes in bone mineral density over time, as well as the interaction with coadministered drugs. Collectively, these studies and analyses served as supportive evidence for the effectiveness of the approved dosages and provided general dosing instructions of the first approved oral gonadotropin-releasing hormone receptor antagonist.


Subject(s)
Endometriosis/drug therapy , Hormone Antagonists/pharmacokinetics , Hydrocarbons, Fluorinated/pharmacokinetics , Organic Anion Transporters/antagonists & inhibitors , Pyrimidines/pharmacokinetics , Receptors, LHRH/antagonists & inhibitors , Administration, Oral , Bone Density/drug effects , Drug Interactions/physiology , Endometriosis/complications , Endometriosis/metabolism , Female , Gonadotropin-Releasing Hormone/drug effects , Hormone Antagonists/administration & dosage , Hormone Antagonists/pharmacology , Humans , Hydrocarbons, Fluorinated/administration & dosage , Hydrocarbons, Fluorinated/pharmacology , Liver Diseases/complications , Organic Anion Transporters/metabolism , Pain/drug therapy , Pain/etiology , Pharmacogenetics , Pharmacology, Clinical , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Treatment Outcome
9.
Gen Comp Endocrinol ; 287: 113342, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31783025

ABSTRACT

Copper is a metal ion present in all organisms, where it has well-known roles in association with proteins and enzymes essential for cellular processes. In the early decades of the twentieth century copper was shown to influence mammalian reproductive biology, and it was subsequently shown to exert effects primarily at the level of the pituitary gland and/or hypothalamic regions of the brain. Furthermore, it has been reported that copper can interact with key neuropeptides in the hypothalamic-pituitary-gonadal axis, notably gonadotropin-releasing hormone (GnRH) and neurokinin B. Interestingly, recent phylogenetic analysis of the sequences of GnRH-related peptides indicates that copper binding is an evolutionarily ancient property of this neuropeptide family, which has been variously retained, modified or lost in the different taxa. In this mini-review the metal-binding properties of neuropeptides in the vertebrate reproductive pathway are reviewed and the evolutionary and functional significance of copper binding by GnRH-related neuropeptides in vertebrates and invertebrates are discussed.


Subject(s)
Copper/pharmacology , Endocrine System/drug effects , Gonadotropin-Releasing Hormone/drug effects , Neurokinin B/drug effects , Reproduction/drug effects , Animals , Endocrine System/physiology , Gonadotropin-Releasing Hormone/chemistry , Gonadotropin-Releasing Hormone/physiology , Invertebrates/metabolism , Mammals/metabolism , Neurokinin B/chemistry , Neurokinin B/physiology , Protein Conformation/drug effects , Reproduction/physiology , Structure-Activity Relationship , Vertebrates/metabolism
10.
Life Sci ; 227: 166-174, 2019 Jun 15.
Article in English | MEDLINE | ID: mdl-31026452

ABSTRACT

AIMS: To investigate the direct histomorphological clues and observe the biological effects of VP acting on gonadotropin-releasing hormone (GnRH) secretion. MAIN METHODS: Immunofluorescence was conducted to investigate the expressions of GnRH and VP in experimental left varicocele (ELV) rats and ELV repair rats. The colocalization of GnRH and VP was observed by electron microscopy immunohistochemistry. The protein-protein interaction between GnRH and VP was tested by co-immunoprecipitation (co-IP) and the proximity ligation assay (PLA). The effects of intracellular and extracellular VP on GnRH and relative transcription factors (Oct-1, Otx2, Pbx1b and DREAM) were respectively evaluated in VP overexpressed and VP treated GT1-7 cells. KEY FINDINGS: Both hypothalamic GnRH and VP decreased in ELV rats and recovered by ELV repair. The overlapped immunolocalizations of GnRH and VP mainly distributed in the lateral part of the arcuate nucleus (ArcL) and median eminence (ME) with a Manders' overlap coefficient of 0.743 ±â€¯0.117. Immunoreactive substances of GnRH and VP existed in the same and adjacent terminals. VP overexpression did not cause any significant effects on the expressions of GnRH and Oct-1, as well as GnRH promoter activity. While 50-200 pg/ml VP treatments increased GnRH mRNA levels in a dose- and time-dependent manner in GT1-7 cells. Additionally, 200 pg/ml VP triggered a marked promotion of expressions of GnRH, Oct-1, Oxt2 Pbx1b and DREAM, as well as GnRH promoter activity (P < 0.05). SIGNIFICANCE: The results reveal the colocalization and interaction of VP and GnRH, which will be conducive to explain the effects and mechanisms of VP acting on reproduction.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Vasopressins/metabolism , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Cell Line , Gonadotropin-Releasing Hormone/drug effects , Gonadotropins/metabolism , Hypothalamus/cytology , Hypothalamus/drug effects , Male , Median Eminence/metabolism , Neurons/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Transcription Factors/metabolism , Vasopressins/pharmacology
11.
Res Vet Sci ; 123: 51-58, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30586652

ABSTRACT

The effects of obestatin on gonadotrophic axis activity in ruminants have not yet been determined. The aim of this study was to investigate the effect of intracerebroventricular infusions of obestatin on the gonadotrophin-releasing hormone (GnRH) mRNA and protein expressions as well as on KNDy mRNA and kisspeptin (Kiss) peptide expressions in peripubertal female sheep. Animals were randomly divided into two groups: the control group received intracerebroventricular infusions of the vehicle, and the obestatin group was infused with obestatin (25 µg/120 µL h-1). The series of four 1-h infusions per day during three consecutive days were performed. After the end of the experiment parts of sheep brains were fixed in situ for immunohistochemical analysis, while the remaining brains were frozen for Real Time qPCR analysis. Substantial changes in the activity of the GnRH and KNDy gene network were observed in obestatin-infused sheep. In those animals an increase of GnRH mRNA expression in the preoptic area, a decrease of GnRH mRNA expression in the median eminence and an increase of GnRH immunoreactivity in the median eminence were found. Moreover, changes in the KNDy mRNA expression in mediobasal hypothalamus as well as decrease Kiss expression in arcuate nucleus and median eminence were observed. It was revealed that obestatin affects the GnRH and KNDy gene network as well as Kiss at the level of mRNA and protein expression. Thereby, it can be concluded that obestatin participates in the mechanism modulating gonadotrophic axis activity at the central level in peripubertal female sheep.


Subject(s)
Ghrelin/pharmacology , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/drug effects , Sheep/physiology , Animals , Female , Gene Expression Regulation/drug effects , Ghrelin/administration & dosage , Ghrelin/metabolism , Gonadotropin-Releasing Hormone/drug effects , Gonadotropin-Releasing Hormone/genetics , Hypothalamus/diagnostic imaging , Hypothalamus/metabolism , Immunohistochemistry , Infusions, Intraventricular , RNA, Messenger/metabolism , Random Allocation , Real-Time Polymerase Chain Reaction , Sexual Maturation
12.
Acta Neurobiol Exp (Wars) ; 78(4): 352-357, 2018.
Article in English | MEDLINE | ID: mdl-30624434

ABSTRACT

It has been reported that gonadotropin­releasing hormone (GnRH), and its analogue leuprolide acetate (LA), have neurotrophic properties; particularly in the regeneration of injured spinal cord in animal models and in the case of a patient with spinal cord injury (SCI). The aim of this study was to establish whether treatment with LA improves sensitivity, motor activity and independence in patients with chronic SCI. Patients were treated LA once a month for six months. They were evaluated at the beginning and at the end of treatment; using a sensitivity and motor impairment scale, according to the American Spinal Injury Association (ASIA), and grade of independence scale; employing the spinal cord independence measure (SCIM). Statistical analysis showed a significant improvement in the ASIA sensory score and the SCIM score when comparing the initial versus final evaluation after six months of LA administration. Some patients showed an increase in frequency of bowel movements. Treatment with LA induces improvements in sensitivity, motor activity and independence in patients with chronic SCI. One advantage of this protocol is that it is a non-invasive method of easy and safe application, with few side effects.


Subject(s)
Gonadotropin-Releasing Hormone/drug effects , Leuprolide/pharmacology , Recovery of Function/drug effects , Spinal Cord Injuries/drug therapy , Adolescent , Adult , Chronic Disease , Female , Gonadotropin-Releasing Hormone/biosynthesis , Humans , Male , Middle Aged , Treatment Outcome , Young Adult
13.
Brain Res ; 1650: 21-30, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27566061

ABSTRACT

The time course effects of ovarian steroids on kisspeptin and GnRH/LH systems is not totally clarified. We investigated the temporal relationship among kisspeptin and GnRH mRNA and kisspeptin content in the preoptic area (POA), GnRH content and release in the medial basal hypothalamus (MBH) and plasma LH levels under different steroid treatments. Ovariectomized rats treated with oil (OVOO), oil plus single dose of estradiol (OVOE), oil plus single dose of progesterone (OVOP), estradiol for 3 days plus oil (OVEO) or estradiol for 3 days plus progesterone (OVEP) were hourly decapitated from 10:00 to 17:00 or had the MBH microdialyzed from 09:00 to 19:00. Estradiol and progesterone acutely increased POA kisspeptin content without altering POA kisspeptin mRNA levels. Short-term exposure to both hormones stimulated MBH GnRH content, although no GnRH/LH surges had occurred. Chronic estradiol-treatment increased both kisspeptin mRNA levels and content in the POA, demonstrating that long exposure to estradiol is required to activate the whole kisspeptin synthesis machinery. This was followed by the peak in the GnRH/LH release. In estradiol-primed rats, progesterone further increased POA kisspeptin content, amplified and advanced GnRH/LH surges, with no additional change on POA kisspeptin mRNA. The data show an estradiol-induced temporal association between kisspeptin increase in the POA and GnRH/LH surges. Interestingly, the classic action of progesterone in amplifying and accelerating the GnRH/LH surges seems to occur by a mechanism which involves POA kisspeptin system.


Subject(s)
Estradiol/metabolism , Gonadotropin-Releasing Hormone/drug effects , Gonadotropin-Releasing Hormone/metabolism , Animals , Estradiol/pharmacology , Female , Gonadotropins , Hypothalamus/drug effects , Hypothalamus, Middle/drug effects , Kisspeptins/drug effects , Kisspeptins/metabolism , Luteinizing Hormone/blood , Ovariectomy , Preoptic Area/drug effects , Progesterone/metabolism , Progesterone/pharmacology , Rats , Rats, Wistar , Spatio-Temporal Analysis
14.
Environ Pollut ; 207: 299-307, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26432752

ABSTRACT

There has been a gradual increase in production and consumption of atrazine (ATR) in agriculture to meet the population rising demands. Female reproduction is necessary for growth and maintenance of population. However, ATR impact on females and particularly ovarian developmental toxicity is less clear. The aim of this study was to define the pathways by which ATR exerted toxic effects on ovarian development of ovary and hypothalamo-pituitary-ovarian (HPO) axis. Female quails were dosed by oral gavage from sexual immaturity to maturity with 0, 50, 250 and 500 mg ATR/kg/d for 45 days. ATR had no effect on mortality but depressed feed intake and growth and influenced the biochemical parameters. Notably, the arrested development of ovaries and oviducts were observed in ATR-exposed quails. The circulating concentrations of E2, P, LH and PRL were unregulated and FSH and T was downregulated in ATR-treated quails. The mRNA expression of GnRH in hypothalamo and LH in pituitary and FSH in ovary was downregulated significantly by ATR exposure and FSH and PRL in pituitary were upregulated. ATR exposure upregulated the level of P450scc, P450arom, 3ß-HSD and 17ß-HSD in ovary and downregulated ERß expression in female quails. However, ATR did not change ERα expression in ovary. This study provides new insights regarding female productive toxicology of ATR exposure. Ovary and oviduct in sexually maturing females were target organs of ATR-induced developmental toxicity. We propose that ATR-induced developmental abnormality of ovary and oviduct is associated with disruption of gonadal hormone balance and HPO axis in female quails.


Subject(s)
Atrazine/toxicity , Coturnix/growth & development , Ovary/drug effects , Oviducts/drug effects , Animals , Body Weight , Female , Gonadal Hormones , Gonadotropin-Releasing Hormone/drug effects , Hypothalamo-Hypophyseal System/drug effects , Ovary/growth & development , Oviducts/growth & development , Pituitary Gland/drug effects , Reproduction/drug effects
15.
Endocrinology ; 156(7): 2563-70, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25853665

ABSTRACT

Bisphenol A (BPA) is an industrial compound with pervasive distribution in the environments of industrialized countries. The U.S. Centers for Disease Control recently found that greater than 90% of Americans carry detectable levels of BPA, raising concern over the direct influences of this compound on human physiology. Epidemiologic evidence links elevated BPA serum concentrations to human reproductive dysfunction, although controlled studies on the acute effect of BPA exposure on reproductive function are limited, particularly in primates. We evaluated the effect of direct BPA exposure on female primate hypothalamic peptide release. Specifically, using a microdialysis method, we examined the effects of BPA (0.1, 1, and 10nM) directly infused to the stalk-median eminence on the release of GnRH and kisspeptin (KP) in mid to late pubertal ovarian intact female rhesus monkeys. We found that the highest level of BPA exposure (10nM) suppressed both GnRH and KP release, whereas BPA at lower concentrations (0.1 and 1nM) had no apparent effects. In addition, we measured BPA in plasma and hypothalamic dialysates after an iv bolus injection of BPA (100 µg/kg). We found a relatively stable distribution of BPA between the blood and brain (plasma:brain ≅ 5:1) persists across a wide range of blood BPA concentrations (1-620 ng/mL). Findings of this study suggest that persistent, high-level exposures to BPA could impair female reproductive function by directly influencing hypothalamic neuroendocrine function.


Subject(s)
Benzhydryl Compounds/pharmacology , Estrogens, Non-Steroidal/pharmacology , Gonadotropin-Releasing Hormone/drug effects , Hypothalamus/drug effects , Kisspeptins/drug effects , Phenols/pharmacology , Animals , Female , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Kisspeptins/metabolism , Macaca mulatta , Median Eminence , Microdialysis , Pituitary Gland
16.
Endocrinology ; 156(5): 1804-14, 2015 May.
Article in English | MEDLINE | ID: mdl-25734362

ABSTRACT

Our recent study indicates that a brief infusion (20 min) of estradiol (E2) benzoate (EB) into the stalk-median eminence (S-ME) stimulates GnRH release with a latency of approximately 10 minutes. In contrast to the effect induced by a brief infusion of EB, it has previously been shown that systemic EB administration suppresses release of GnRH, kisspeptin, and LH with a latency of several hours, which is known as the negative feedback action of E2. We speculated that the differential results by these 2 modes of EB administration are due to the length of E2 exposure. Therefore, in the present study, the effects of EB infusion for periods of 20 minutes, 4 hours, or 7 hours into the S-ME of ovariectomized female monkeys on the release of GnRH and kisspeptin were examined using a microdialysis method. To assess the effects of the EB infusion on LH release, serum samples were also collected. The results show that similar to the results with 20-minute infusion, both 4- and 7-hour infusions of EB consistently stimulated release of GnRH and kisspeptin from the S-ME accompanied by LH release in the general circulation. In contrast, sc injection of EB suppressed all 3 hormones (GnRH, kisspeptin, and LH) measured. It is concluded that regardless of the exposure period, direct E2 action on GnRH and kisspeptin neurons in the S-ME, where their neuroterminals are present, is stimulatory, and the E2-negative feedback effects do not occur at the S-ME level.


Subject(s)
Contraceptive Agents/pharmacology , Estradiol/analogs & derivatives , Gonadotropin-Releasing Hormone/drug effects , Kisspeptins/drug effects , Median Eminence/drug effects , Animals , Contraceptive Agents/administration & dosage , Estradiol/administration & dosage , Estradiol/pharmacology , Female , Gonadotropin-Releasing Hormone/metabolism , Kisspeptins/metabolism , Macaca mulatta , Median Eminence/metabolism , Microdialysis , Ovariectomy
17.
Endocrinology ; 156(5): 1828-37, 2015 May.
Article in English | MEDLINE | ID: mdl-25710282

ABSTRACT

We tested the hypothesis that kisspeptin cells constitute the "pulse generator" for GnRH secretion. In ewes, we determined whether iv administered kisspeptin elicits a secretory pulse of LH in anaesthetized, sex-steroid suppressed ovariectomized ewes. A response was seen in both anaesthetized and conscious animals, which was not associated with induction of c-Fos labeling in GnRH cells, supporting the notion that kisspeptin acts on the neurosecretory GnRH terminals. Response was lower in the anaesthetized animals, suggesting that some nonkisspeptin elements may be involved in GnRH responses. Microinjection of kisspeptin (100 nmol) into the median eminence of conscious ewes elicited a pulse of LH, indicating that kisspeptin acts at this level to cause GnRH secretion. To determine which cells are activated at the time of GnRH secretion, we blood sampled 18 ewes during the luteal phase of the estrous cycle and harvested brains after 3 hours. Three of these ewes displayed a pulse of LH within 30 minutes of euthanasia. An increase in c-Fos labeling was seen in kisspeptin and glutamate cells of the arcuate nucleus but not in GnRH neurons, preoptic kisspeptin neurons, or preoptic glutamate neurons. Immunohistochemistry in 4 hypothalami showed that 72% of arcuate kisspeptin cells receive glutamatergic input. These data support the concept that the kisspeptin cells of the arcuate nucleus drive pulsatile secretion of GnRH at the level of the median eminence, but this may involve "upstream" input from glutamate cells. We conclude that the pulse generator for GnRH secretion involves more than 1 element.


Subject(s)
Arcuate Nucleus of Hypothalamus/drug effects , Gonadotropin-Releasing Hormone/drug effects , Kisspeptins/pharmacology , Luteinizing Hormone/drug effects , Median Eminence/drug effects , Neuroendocrine Cells/drug effects , Periodicity , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Female , Glutamic Acid/metabolism , Gonadotropin-Releasing Hormone/metabolism , Kisspeptins/physiology , Luteinizing Hormone/metabolism , Median Eminence/metabolism , Neuroendocrine Cells/metabolism , Neurons/drug effects , Neurons/metabolism , Ovariectomy , Proto-Oncogene Proteins c-fos/metabolism , Sheep
18.
Neuro Endocrinol Lett ; 36(8): 767-70, 2015.
Article in English | MEDLINE | ID: mdl-26921577

ABSTRACT

OBJECTIVE: The neonatal and/or prepubertal androgen milieu affects sexual maturation. In rodents, neonatal chronic testosterone treatment, which is used as a model of polycystic ovary syndrome (PCOS), results in the onset of vaginal opening occurring earlier in the pubertal period. DESIGN: In the present study, the changes in hypothalamic Kiss1 (a gonadotropin-releasing hormone (GnRH)-stimulating factor) and RF-amide related peptide (RFRP; a GnRH inhibitory factor) mRNA expression induced by testosterone treatment were examined in order to clarify whether these factors are involved in the testosterone-induced acceleration of sexual maturation. RESULTS: The onset of vaginal opening occurred earlier and uterine weight was increased in female rats subjected to chronic (from postnatal day 23 to day 31) testosterone treatment. Contrary to our expectations, the rats' hypothalamic Kiss1 and Kiss1 receptor mRNA levels were not changed, and their serum luteinizing hormone (LH) levels were decreased. Although hypothalamic RFRP mRNA expression was decreased in the testosterone-treated rats, this change was not reflected in their serum LH levels. CONCLUSIONS: These results indicate that the advancement of sexual maturation observed in chronic testosterone-treated rats might be caused by a peripheral, rather than a central, mechanism.


Subject(s)
Androgens/pharmacology , Hypothalamus/drug effects , Kisspeptins/drug effects , Neuropeptides/drug effects , RNA, Messenger/drug effects , Sexual Maturation/drug effects , Testosterone/pharmacology , Vagina/drug effects , Animals , Female , Gonadotropin-Releasing Hormone/drug effects , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Kisspeptins/genetics , Kisspeptins/metabolism , Neuropeptides/genetics , Neuropeptides/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Kisspeptin-1 , Receptors, Neuropeptide/drug effects , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism
19.
JSLS ; 18(3)2014.
Article in English | MEDLINE | ID: mdl-25392665

ABSTRACT

BACKGROUND AND OBJECTIVES: The aim of this study was to compare the in vitro fertilization (IVF) outcomes of long gonadotropin-releasing hormone agonist (GnRH-a) and GnRH-antagonist (GnRH-ant) protocols in endometriosis patients who have undergone laparoscopic endometrioma resection surgery. To our knowledge, there is no study in the current literature that compares the effectiveness of long GnRH-a and GnRH-ant protocols in management of IVF cycles in endometriosis patients who underwent laparoscopic endometrioma resection surgery. METHODS: Eighty-six patients with stage III to IV endometriosis who had undergone laparoscopic resection surgery for endometrioma were divided into 2 groups: those who had ovarian stimulation with a long GnRH-a protocol (n=44), and those who had ovarian stimulation with a GnRH-ant protocol (n=42). RESULTS: The number of follicles on human chorionic gonadotropin injection day, duration of hyperstimulation, number of retrieved metaphase II oocytes, and total number of grade 1 embryos were statically significantly higher in the long GnRH-a protocol. There were no significant differences in positive ß-human chorionic gonadotropin pregnancy rates (25% vs 21.4%; P=.269) and ongoing pregnancy rates per patient (20.5% vs 19.1%; P=.302) between the 2 protocols. CONCLUSIONS: Long GnRH-a and GnRH-ant protocols both present similar IVF outcomes in patients with endometriosis who have undergone laparoscopic endometrioma resection surgery. A long GnRH-a protocol may lead to a higher number of embryos that can be cryopreserved, providing the possibility of additional embryo transfers without having to go through the process of ovarian stimulation again.


Subject(s)
Endometriosis/surgery , Fertilization in Vitro/methods , Gonadotropin-Releasing Hormone/drug effects , Leuprolide/therapeutic use , Ovulation Induction/methods , Postoperative Care/methods , Triptorelin Pamoate/therapeutic use , Adolescent , Adult , Endometrial Neoplasms/surgery , Female , Fertility Agents, Female/therapeutic use , Gonadotropin-Releasing Hormone/metabolism , Humans , Luteolytic Agents/therapeutic use , Pregnancy , Pregnancy Rate/trends , Young Adult
20.
Minerva Ginecol ; 66(2): 133-53, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24848073

ABSTRACT

Premature ovarian insufficiency, defined as amenorrhea with estrogen deficiency in a woman younger than 40 associated with a serum follicle stimulating hormone (FSH) >35 mIU/mL, can be temporarily reversed with ovulation achieved resulting in live delivered pregnancies. Though this may occur spontaneously the frequency of ovulation can be considerably increased by various techniques of lowering the elevated serum FSH level and thus up-regulate down-regulated FSH receptors in the granulosa-theca cells. This can be accomplished by either suppressing FSH release from the pituitary by negative feedback through high dose estrogen or by suppressing FSH production by inhibiting the gonadotropin releasing hormone (GnRH) by either using GnRH agonists or antagonists. The estrogen method is the technique of choice because it is much less expensive than GnRH analogues, and helps stimulate cervical mucus and endometrial development. Ethinyl estradiol is the preferred estrogen because it does not contribute to the measurement of serum estradiol and thus allows proper monitoring of follicular maturation. Sometimes exogenous gonadotropins are needed as a boost but the dosage should be low so as not to down-regulate FSH receptors again. The technique is referred to as the FSH receptor restoration technique. Progesterone should be supplemented in the luteal phase. Physicians should be cognizant of trying to help prevent premature ovarian insufficiency by judiciously choosing less gonadotoxic cancer treatment alternatives that are equally efficacious. Also surgery for ovarian endometriomas should be performed only when absolutely necessary.


Subject(s)
Follicle Stimulating Hormone/blood , Infertility, Female/therapy , Primary Ovarian Insufficiency/complications , Adult , Animals , Estrogens/administration & dosage , Estrogens/deficiency , Ethinyl Estradiol/administration & dosage , Ethinyl Estradiol/therapeutic use , Female , Gonadotropin-Releasing Hormone/drug effects , Gonadotropin-Releasing Hormone/metabolism , Gonadotropins/administration & dosage , Gonadotropins/therapeutic use , Humans , Infertility, Female/etiology , Ovulation/physiology , Primary Ovarian Insufficiency/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...