Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Arch. endocrinol. metab. (Online) ; 63(4): 438-444, July-Aug. 2019. tab, graf
Article in English | LILACS | ID: biblio-1019366

ABSTRACT

ABSTRACT Pubertal timing in humans is determined by complex interactions including hormonal, metabolic, environmental, ethnic, and genetic factors. Central precocious puberty (CPP) is defined as the premature reactivation of the hypothalamic-pituitary-gonadal axis, starting before the ages of 8 and 9 years in girls and boys, respectively; familial CPP is defined by the occurrence of CPP in two or more family members. Pioneering studies have evidenced the participation of genetic factors in pubertal timing, mainly identifying genetic causes of CPP in sporadic and familial cases. In this context, rare activating mutations were identified in genes of the kisspeptin excitatory pathway (KISS1R and KISS1 mutations). More recently, loss-of-function mutations in two imprinted genes (MKRN3 and DLK1) have been identified as important causes of familial CPP, describing novel players in the modulation of the hypothalamic-pituitary-gonadal axis in physiological and pathological conditions. MKRN3 mutations are the most common cause of familial CPP, and patients with MKRN3 mutations present clinical features indistinguishable from idiopathic CPP. Meanwhile, adult patients with DLK1 mutations present high frequency of metabolic alterations (overweight/obesity, early onset type 2 diabetes and hyperlipidemia), indicating that DLK1 may be a novel link between reproduction and metabolism. Arch Endocrinol Metab. 2019;63(4):438-44


Subject(s)
Humans , Puberty, Precocious/genetics , Phenotype , Puberty, Precocious/etiology , Ribonucleoproteins/genetics , Calcium-Binding Proteins , Gene Silencing , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Kisspeptins/genetics , Receptors, Kisspeptin-1/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Methylation , Mutation
2.
Clinics ; 72(8): 510-514, Aug. 2017. tab, graf
Article in English | LILACS | ID: biblio-890718

ABSTRACT

OBJECTIVES: Polycystic ovary syndrome is a heterogeneous endocrine disorder that affects reproductive-age women. The mechanisms underlying the endocrine heterogeneity and neuroendocrinology of polycystic ovary syndrome are still unclear. In this study, we investigated the expression of the kisspeptin system and gonadotropin-releasing hormone pulse regulators in the hypothalamus as well as factors related to luteinizing hormone secretion in the pituitary of polycystic ovary syndrome rat models induced by testosterone or estradiol. METHODS: A single injection of testosterone propionate (1.25 mg) (n=10) or estradiol benzoate (0.5 mg) (n=10) was administered to female rats at 2 days of age to induce experimental polycystic ovary syndrome. Controls were injected with a vehicle (n=10). Animals were euthanized at 90-94 days of age, and the hypothalamus and pituitary gland were used for gene expression analysis. RESULTS: Rats exposed to testosterone exhibited increased transcriptional expression of the androgen receptor and estrogen receptor-β and reduced expression of kisspeptin in the hypothalamus. However, rats exposed to estradiol did not show any significant changes in hormone levels relative to controls but exhibited hypothalamic downregulation of kisspeptin, tachykinin 3 and estrogen receptor-α genes and upregulation of the gene that encodes the kisspeptin receptor. CONCLUSIONS: Testosterone- and estradiol-exposed rats with different endocrine phenotypes showed differential transcriptional expression of members of the kisspeptin system and sex steroid receptors in the hypothalamus. These differences might account for the different endocrine phenotypes found in testosterone- and estradiol-induced polycystic ovary syndrome rats.


Subject(s)
Animals , Female , Gonadotropin-Releasing Hormone/analysis , Hypothalamus/chemistry , Kisspeptins/analysis , Luteinizing Hormone/metabolism , Pituitary Gland/metabolism , Polycystic Ovary Syndrome/chemistry , Disease Models, Animal , Down-Regulation , Estradiol , Gene Expression , Gonadotropin-Releasing Hormone/genetics , Hypothalamus/metabolism , Kisspeptins/genetics , Phenotype , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism , Rats, Wistar , Real-Time Polymerase Chain Reaction , Receptors, Androgen/analysis , Receptors, Estrogen/analysis , Testosterone , Up-Regulation
3.
Arch. endocrinol. metab. (Online) ; 60(6): 587-595, Nov.-Dec. 2016. tab, graf
Article in English | LILACS | ID: biblio-827788

ABSTRACT

ABSTRACT Prolactin is best known for its effects of stimulating mammary gland development and lactogenesis. However, prolactin is a pleiotropic hormone that is able to affect several physiological functions, including fertility. Prolactin receptors (PRLRs) are widely expressed in several tissues, including several brain regions and reproductive tract organs. Upon activation, PRLRs may exert prolactin’s functions through several signaling pathways, although the recruitment of the signal transducer and activator of transcription 5 causes most of the known effects of prolactin. Pathological hyperprolactinemia is mainly due to the presence of a prolactinoma or pharmacological effects induced by drugs that interact with the dopamine system. Notably, hyperprolactinemia is a frequent cause of reproductive dysfunction and may lead to infertility in males and females. Recently, several studies have indicated that prolactin may modulate the reproductive axis by acting on specific populations of hypothalamic neurons that express the Kiss1 gene. The Kiss1 gene encodes neuropeptides known as kisspeptins, which are powerful activators of gonadotropin-releasing hormone neurons. In the present review, we will summarize the current knowledge about prolactin’s actions on reproduction. Among other aspects, we will discuss whether the interaction between prolactin and the Kiss1-expressing neurons can affect reproduction and how kisspeptins may become a novel therapeutic approach to treat prolactin-induced infertility.


Subject(s)
Humans , Male , Female , Prolactin/metabolism , Reproduction/physiology , Kisspeptins/metabolism , Prolactin/pharmacology , Receptors, Prolactin/metabolism , Hyperprolactinemia/complications , Signal Transduction , Sex Factors , Hypothalamus/metabolism , Infertility/etiology
4.
Journal of Southern Medical University ; (12): 532-537, 2016.
Article in Chinese | WPRIM | ID: wpr-264009

ABSTRACT

<p><b>OBJECTIVE</b>To identify potential markers for predicting invasion, metastasis, and prognosis of gastric adenocarcinoma (GAC).</p><p><b>METHODS</b>The expressions of Slug, ZEB1 and KISS-1 were detected immunohistochemically in 261 GAC tissues and 80 normal gastric tissues.</p><p><b>RESULTS</b>The positivity rates of Slug, ZEB1, and KISS-1 in gastric tissues were 2.5%, 1.3%, and 87.5%, respectively, significantly different from the rates of 62.1%, 28.4%, and 41.1% in GAC tissues (P<0.05). The expression level of Slug was significantly correlated with the depth of invasion, lymph node metastasis, and pTNM stages; the positivity rates of both ZEB1 and KISS-1 were significantly correlated with the tumor grade, depth of invasion, lymph node metastasis and pTNM stages. Slug expression was positively correlated with ZEB1 expression, and KISS-1 expression was inversely correlated with Slug and ZEB1 expressions. Kaplan-Meier analysis showed that the overall survival time of patients with positive expressions of Slug and ZEB1 was significantly shorter than that of the negative patients, and the survival time of patients positive for KISS-1 was significantly longer than the negative patients. COX multivariate analysis showed that positive Slug, ZEB1 and KISS-1 protein expressions and pTNM stages were independent prognostic factors of GAC (P<0.05).</p><p><b>CONCLUSION</b>The abnormal expressions of Slug, ZEB1 and KISS-1 may contribute to the tumorigenesis of GAC and are related with lymph node metastasis, pTNM stages, and prognosis of GAC. The combined detection of Slug, ZEB1, and KISS-1 expression has an important value in predicting the progression and prognosis of GAC.</p>


Subject(s)
Humans , Adenocarcinoma , Metabolism , Pathology , Disease Progression , Homeodomain Proteins , Metabolism , Immunohistochemistry , Kaplan-Meier Estimate , Kisspeptins , Metabolism , Lymphatic Metastasis , Neoplasm Grading , Prognosis , Proportional Hazards Models , Snail Family Transcription Factors , Stomach Neoplasms , Metabolism , Pathology , Transcription Factors , Metabolism , Zinc Finger E-box-Binding Homeobox 1
5.
Annals of Pediatric Endocrinology & Metabolism ; : 66-69, 2016.
Article in English | WPRIM | ID: wpr-145165

ABSTRACT

Central precocious puberty (CPP) is caused by the premature reactivation of the hypothalamic-pituitary-gonadal axis. Genetic, nutritional, and environmental factors play a crucial role in determining pubertal timing. Recently mutations in kisspeptin (KISS1), kisspeptin receptor (KISS1R), and makorin RING finger protein 3 (MKRN3) genes have been identified as genetic causes of CPP. In particular, the MKRN3 gene is known to affect pubertal initiation. The MKRN3 gene is located on chromosome 15q11-q13 in the Prader-Willi syndrome (PWS) critical region. MKRN3 deficiency, due to a loss of function mutation, leads to the withdrawal of hypothalamic inhibition and prompts pulsatile gonadotropin-releasing hormone secretion, resulting in precocious puberty. The exact functions of these genes associated with CPP are still not well understood. Larger studies are required to discover the mechanisms involved in pubertal development.


Subject(s)
Fingers , Gonadotropin-Releasing Hormone , Kisspeptins , Prader-Willi Syndrome , Puberty, Precocious
6.
Annals of Pediatric Endocrinology & Metabolism ; : 66-69, 2016.
Article in English | WPRIM | ID: wpr-145152

ABSTRACT

Central precocious puberty (CPP) is caused by the premature reactivation of the hypothalamic-pituitary-gonadal axis. Genetic, nutritional, and environmental factors play a crucial role in determining pubertal timing. Recently mutations in kisspeptin (KISS1), kisspeptin receptor (KISS1R), and makorin RING finger protein 3 (MKRN3) genes have been identified as genetic causes of CPP. In particular, the MKRN3 gene is known to affect pubertal initiation. The MKRN3 gene is located on chromosome 15q11-q13 in the Prader-Willi syndrome (PWS) critical region. MKRN3 deficiency, due to a loss of function mutation, leads to the withdrawal of hypothalamic inhibition and prompts pulsatile gonadotropin-releasing hormone secretion, resulting in precocious puberty. The exact functions of these genes associated with CPP are still not well understood. Larger studies are required to discover the mechanisms involved in pubertal development.


Subject(s)
Fingers , Gonadotropin-Releasing Hormone , Kisspeptins , Prader-Willi Syndrome , Puberty, Precocious
7.
Journal of Southern Medical University ; (12): 1643-1648, 2015.
Article in Chinese | WPRIM | ID: wpr-232554

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the effect of Kiss-1 gene suppression on the metastatic capacity of HCT116 human colorectal carcinoma cells in vitro and the involvement of nuclear factor-κB (NF-κB) signaling pathway.</p><p><b>METHODS</b>A recombinant lentiviral vector of Kiss-1 gene pGC-LV-Kiss-1-EGFP or the empty vector was transfected in HCT116 cells. Cell Counting Kit-8 (CCK8) and Transwell chamber assay were used to detect the changes in cell proliferation, invasion and migration ability after the transfection. Western blotting was used to detect the expression of I-κB, the inhibitive protein of NF-κB signal pathway, and the expression of the downstream effector MMP-9 before and after transfection.</p><p><b>RESULTS</b>In cells over-expressing Kiss-1, I-κB expression increased and MMP-9 expression decreased significantly compared to those in the blank control and vector-transfected cells (P<0.05). Kiss-1 gene over-expression resulted in significant inhibition of HCT116 cell proliferation, invasion, and migration as compared to the control cells (P<0.05).</p><p><b>CONCLUSION</b>Lentivirus-mediated Kiss-1 gene over-expression can inhibit the proliferation, invasion, and migration of HCT116 cells via the NF-B signaling pathway.</p>


Subject(s)
Humans , Cell Movement , Cell Proliferation , Colorectal Neoplasms , Pathology , Genetic Vectors , HCT116 Cells , I-kappa B Kinase , Metabolism , Kisspeptins , Genetics , Lentivirus , Matrix Metalloproteinase 9 , Metabolism , NF-kappa B , Metabolism , Neoplasm Invasiveness , Signal Transduction , Transfection
8.
Endocrinology and Metabolism ; : 124-141, 2015.
Article in English | WPRIM | ID: wpr-30202

ABSTRACT

Kisspeptin has recently emerged as a key regulator of the mammalian reproductive axis. It is known that kisspeptin, acting centrally via the kisspeptin receptor, stimulates secretion of gonadotrophin releasing hormone (GnRH). Loss of kisspeptin signaling causes hypogonadotrophic hypogonadism in humans and other mammals. Kisspeptin interacts with other neuropeptides such as neurokinin B and dynorphin, to regulate GnRH pulse generation. In addition, a growing body of evidence suggests that kisspeptin signaling be regulated by nutritional status and stress. Kisspeptin may also represent a novel potential therapeutic target in the treatment of fertility disorders. Early human studies suggest that peripheral exogenous kisspeptin administration stimulates gonadotrophin release in healthy adults and in patients with certain forms of infertility. This review aims to concisely summarize what is known about kisspeptin as a regulator of reproductive function, and provide an update on recent advances within this field.


Subject(s)
Adult , Humans , Dynorphins , Fertility , Gonadotropin-Releasing Hormone , Hypogonadism , Hypothalamus , Infertility , Kisspeptins , Mammals , Neurokinin B , Neuropeptides , Nutritional Status , Axis, Cervical Vertebra
9.
Singapore medical journal ; : 649-656, 2015.
Article in English | WPRIM | ID: wpr-276736

ABSTRACT

Kisspeptins are a group of peptide fragments encoded by the KISS1 gene in humans. They bind to kisspeptin receptors with equal efficacy. Kisspeptins and their receptors are expressed by neurons in the arcuate and anteroventral periventricular nuclei of the hypothalamus. Oestrogen mediates negative feedback of gonadotrophin-releasing hormone secretion via the arcuate nucleus. Conversely, it exerts positive feedback via the anteroventral periventricular nucleus. The sexual dimorphism of these nuclei accounts for the differential behaviour of the hypothalamic-pituitary-gonadal axis between genders. Kisspeptins are essential for reproductive function. Puberty is regulated by the maturation of kisspeptin neurons and by interactions between kisspeptins and leptin. Hence, kisspeptins have potential diagnostic and therapeutic applications. Kisspeptin agonists may be used to localise lesions in cases of hypothalamic-pituitary-gonadal axis dysfunction and evaluate the gonadotrophic potential of subfertile individuals. Kisspeptin antagonists may be useful as contraceptives in women, through the prevention of premature luteinisation during in vitro fertilisation, and in the treatment of sex steroid-dependent diseases and metastatic cancers.


Subject(s)
Animals , Female , Humans , Male , Mice , Rats , Arcuate Nucleus of Hypothalamus , Metabolism , Estrogens , Metabolism , Feedback, Physiological , Fertilization in Vitro , Gonadotropin-Releasing Hormone , Metabolism , Homeostasis , Kisspeptins , Physiology , Neoplasms , Metabolism , Neurons , Metabolism , Protein Binding , Reproduction , Sex Factors , Signal Transduction
10.
IJFS-International Journal of Fertility and Sterility. 2014; 8 (3): 333-340
in English | IMEMR | ID: emr-148949

ABSTRACT

RFamide-related peptide-3 [RFRP-3] and kisspeptin [KiSS-1] are known to respectively inhibit and stimulate gonadotropin releasing hormone [GnRH] and luteinizing hormone [LH] secretion in rat. The aim of the present study was to evaluate the relative mRNA expression of RFRP-3 and KiSS-1 in the hypothalamus of pregnant rats. In a randomized controlled experimental study, the exact pregnancy day of 18 Sprague-Dawley rats were confirmed using the vaginal smear method and were equally assigned to three groups of days 7, 14 and 21 of pregnancy. Four non-pregnant female rats were ovariectomized and assigned as the control group. All rats were decapitated, and the dorsomedial hypothalamic nucleus [DMH] and the arcuate nucleus [ARC] for detection of KiSS-1 mRNA were separated from their hypothalamus to detect RFRP-3 and KiSS-1 mRNA respectively. Then, their relative expressions were compared between control and pregnant groups using real-time polymerase chain reaction [PCR]. The relative expression of RFRP-3 mRNA in DMH did not change significantly during pregnancy [p>0.01]. However, the relative expression of KiSS-1 mRNA in ARC was at its highest in day 7 of pregnancy and decreased until day 21 of pregnancy [p<0.01]. Decrease in GnRH and LH secretion during the pregnancy of rat may be controlled by constant expression of RFRP-3 mRNA and reduced expression of KiSS-1 mRNA in hypothalamus


Subject(s)
Animals, Laboratory , RNA, Messenger , Dorsomedial Hypothalamic Nucleus , Kisspeptins , Arcuate Nucleus of Hypothalamus , Rats, Sprague-Dawley , Pregnancy
11.
Chinese Journal of Contemporary Pediatrics ; (12): 754-758, 2014.
Article in Chinese | WPRIM | ID: wpr-254207

ABSTRACT

<p><b>OBJECTIVE</b>To evaluate the effects of neonatal exposure to different doses of bisphenol A (BPA) on the vaginal opening day (VOD), hypothalamic Kiss-1 mRNA expression, and ovarian estrogen receptor (ER) mRNA expression in female rats.</p><p><b>METHODS</b>Neonatal female Sprague-Dawley (SD) rats were randomly divided into six groups: blank control, vehicle, 17β-estradiol (17β-estradiol, E2, 10 μg/d), low-dose BPA [25 μg(kg·d)], medium-dose BPA [50 μg(kg·d)], and high-dose BPA groups [250 μg(kg·d)]. The rats were subcutaneously injected with respective agents on postnatal days 0-6. The VOD was recorded, and each rat was sacrificed on the same day. The hypothalamus and ovary were taken and weighed, and the organ coefficients of hypothalamus and ovary were calculated. The hypothalamic Kiss-1 mRNA expression and ovarian ERα and ERβ mRNA expression were measured by real-time PCR.</p><p><b>RESULTS</b>Compared with the control group, the E2 and medium- and high-dose BPA groups had advanced VOD, and the E2 group had significantly reduced hypothalamic Kiss-1 mRNA expression and ovarian ERβ mRNA expression (P<0.05).</p><p><b>CONCLUSIONS</b>Neonatal exposure to medium- and high-dose BPA[50 and 250 μg/(kg·d)] can induce precocious puberty in rats, but it may not result from the change in hypothalamic Kiss-1 mRNA expression. Neonatal exposure to low-dose BPA [25 μg/(kg·d)] does not induce precocious puberty in rats.</p>


Subject(s)
Animals , Female , Male , Rats , Aging , Animals, Newborn , Benzhydryl Compounds , Toxicity , Dose-Response Relationship, Drug , Hypothalamus , Metabolism , Kisspeptins , Genetics , Phenols , Toxicity , Rats, Sprague-Dawley , Receptors, Estrogen , Genetics , Sexual Maturation
12.
National Journal of Andrology ; (12): 792-797, 2014.
Article in Chinese | WPRIM | ID: wpr-309639

ABSTRACT

<p><b>OBJECTIVE</b>To explore the expressions and functions of the kisspeptin/kiss1r system and GnRH in the hypothalamic arcuate nucleus (HAN) and the influence of the kisspeptin/kiss1r system on the hypothalamic-pituitary-testis (HPT) axis in the rat models of diet-induced obesity.</p><p><b>METHODS</b>Ninety newborn SD male rats were randomly assigned to receive normal diet (n = 30) and high-fat diet (n = 60) for the establishment of obesity models. The model rats were again equally divided into a control group and an experimental group, the latter injected with kisspeptin via the lateral ventricle. Then the body mass index (BMI) and endocrine hormone levels of the rats were recorded, the protein expressions of LepR, kisspeptin, kiss1r, and GnRH in the HAN determined by immunohistochemistry and Western blot, and the levels of GnRH mRNA in the HAN measured by qRT-PCR.</p><p><b>RESULTS</b>Significantly increased BMI and hormone levels indicated the successful establishment of diet-induced obesity models. Compared with the normal rats, the protein expressions of LepR, kisspeptin, and GnRH in the HAN were markedly decreased in the controls, and that of GnRH and the levels of LH and T significantly increased, but the expressions of LepR and kiss1r showed no remarkable changes in the experimental rats.</p><p><b>CONCLUSION</b>Lateral ventricular injection of kisspeptin can upregulate obesity-induced low expression of GnRH, correct the dysfunction of the HPT axis, and thus improve reproductive function in rats.</p>


Subject(s)
Animals , Female , Male , Pregnancy , Rats , Arcuate Nucleus of Hypothalamus , Metabolism , Diet, High-Fat , Disease Models, Animal , Gonadotropin-Releasing Hormone , Metabolism , Hypothalamo-Hypophyseal System , Kisspeptins , Metabolism , Obesity , Metabolism , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled , Metabolism , Receptors, Kisspeptin-1 , Receptors, Leptin , Metabolism
13.
Journal of Korean Medical Science ; : 1120-1125, 2014.
Article in English | WPRIM | ID: wpr-141031

ABSTRACT

Kisspeptin/G-protein couple receptor-54 (GPR54) system plays a key role in the activation of the gonadotropic axis at puberty. Central precocious puberty (CPP) is caused by the premature activation of hypothalamic gonadotropin-releasing hormone secretion. This study was aimed to identify KISS1 gene variations and to investigate the associations between KISS1 gene variations and CPP in Korean girls. All coding exons of KISS1 gene were sequenced in Korean girls with CPP (n = 143) and their healthy controls (n = 101). Nine polymorphisms were identified in KISS1 gene. A novel single-nucleotide polymorphism (SNP), 55648176 T/G, was identified for the first time. SNP 55648184 C/G and 55648186 -/T were detected more frequently in CPP group than in control group. SNP 55648176 T/G was detected less frequently in CPP group than in control group. Haplotype GGGC-ACCC was detected less frequently in CPP group. The genetic variations of KISS1 gene can be contributing factors of development of CPP. The association between the gene variations and CPP should be validated by further evidence obtained from large-scaled and functional studies.


Subject(s)
Child , Female , Humans , Base Sequence , Genetic Markers/genetics , Genetic Predisposition to Disease/epidemiology , Kisspeptins/genetics , Molecular Sequence Data , Point Mutation/genetics , Polymorphism, Single Nucleotide/genetics , Prevalence , Puberty, Precocious/epidemiology , Reproducibility of Results , Republic of Korea/epidemiology , Risk Assessment , Sensitivity and Specificity
14.
Journal of Korean Medical Science ; : 1120-1125, 2014.
Article in English | WPRIM | ID: wpr-141030

ABSTRACT

Kisspeptin/G-protein couple receptor-54 (GPR54) system plays a key role in the activation of the gonadotropic axis at puberty. Central precocious puberty (CPP) is caused by the premature activation of hypothalamic gonadotropin-releasing hormone secretion. This study was aimed to identify KISS1 gene variations and to investigate the associations between KISS1 gene variations and CPP in Korean girls. All coding exons of KISS1 gene were sequenced in Korean girls with CPP (n = 143) and their healthy controls (n = 101). Nine polymorphisms were identified in KISS1 gene. A novel single-nucleotide polymorphism (SNP), 55648176 T/G, was identified for the first time. SNP 55648184 C/G and 55648186 -/T were detected more frequently in CPP group than in control group. SNP 55648176 T/G was detected less frequently in CPP group than in control group. Haplotype GGGC-ACCC was detected less frequently in CPP group. The genetic variations of KISS1 gene can be contributing factors of development of CPP. The association between the gene variations and CPP should be validated by further evidence obtained from large-scaled and functional studies.


Subject(s)
Child , Female , Humans , Base Sequence , Genetic Markers/genetics , Genetic Predisposition to Disease/epidemiology , Kisspeptins/genetics , Molecular Sequence Data , Point Mutation/genetics , Polymorphism, Single Nucleotide/genetics , Prevalence , Puberty, Precocious/epidemiology , Reproducibility of Results , Republic of Korea/epidemiology , Risk Assessment , Sensitivity and Specificity
15.
IJFS-International Journal of Fertility and Sterility. 2013; 6 (4): 304-309
in English | IMEMR | ID: emr-140395

ABSTRACT

Kisspeptin and RFamide-related peptide-3 [RFRP-3] are known to affect GnRH/luteinizing hormone [LH] in several species, including the rat. It has been hypothesized that GnRH/LH changes during the rat estrous cycle may result from changes in the expression of KiSS1 and RFRP-3 genes. Therefore, the present study investigates KiSS1 and RFRP-3 gene expression at the transcriptional level in the rat hypothalamus during the estrous cycle. In the present experimental study, 36 adult female Sprague-Dawley rats [3-4 months old] were used to study the expression of KiSS1 and RFRP-3 mRNA in the hypothalamus during the estrous cycle. Four rats were ovariectomized, whereas the remainder were allotted to four different phases of the estrous cycle [n=8 per estrus phase]. Rats were decapitated, and the hypothalami were immediately dissected and frozen in liquid nitrogen. Expressions of KiSS1 and RFRP-3 mRNAs were analyzed by real-time PCR. The expression of KiSS1 mRNA during estrus was lower than other phases of the cycle [p<0.01]. Expression of KiSS1 mRNA during the metestrus phase was lower than the proestrus phase [p<0.01]. The expression of RFRP-3 mRNA during proestrus was lower than the diestrus phase [p<0.01]. Results of the present study showed the role of coordinated expression of KiSS1 and RFRP-3 mRNA in the hypothalamus in the control of the rat estrous cycle


Subject(s)
Female , Animals, Laboratory , Kisspeptins , Neuropeptides , RNA, Messenger , Estrous Cycle , Rats, Sprague-Dawley , Gene Expression , Real-Time Polymerase Chain Reaction
16.
China Journal of Chinese Materia Medica ; (24): 386-390, 2013.
Article in Chinese | WPRIM | ID: wpr-346811

ABSTRACT

<p><b>OBJECTIVE</b>To study the therapeutic effect of Dabuyin Wan on true precocious puberty of female rats and its possible mechanism.</p><p><b>METHOD</b>Twenty-two-day-old female SD rats were subcutaneously injected with 40 mg x kg(-1) N-methyl-DL-aspartic acid (NMA) at 14:00 and 16:00 every day; meanwhile, the rats were given Dabuyin Wan for intervention. Visual inspection was conducted for the time of vaginal opening. The first estrus was observed by yaginal smear test. Their ovaries and uterus were weighed to calculate organ coefficients. Conventional pathological slices were made to observe morphological changes in ovaries and uterus and calculate the thickness of uterine walls and the number of corpus luteums. The level of E2 in serum was detected to assess the therapeutic effect of Dabuyin Wan on NMA precocious puberty in rats. expressions of GnRH, GPR54 and Kiss-1 mRNA in hypothalamus were measured by semi-quantitative RT-PCR to investigate the possible mechanism of Dabuyin Wan.</p><p><b>RESULT</b>Dabuyin Wan at 3.24 g x kg(-1) and 1.62 g x kg(-1) significantly decreased the organ coefficients in rats with precocious puberty (P < 0.05), decrease the number of vaginal openings in rats (P < 0.01) and the thickness of uterine walls and the number of corpus luteums (P < 0.05), and notably down-regulated expressions of GnRH, GPR54 and Kiss-1 mRNA in hypothalamus (P < 0.05), without significant impact on E2 in serum.</p><p><b>CONCLUSION</b>Dabuyin Wan may inhibit GnRH synthesis and release as well as startup of hypothalamic-pituitary-gonadal axis by down-regulating Kiss-1/GPR54 mRNA expression in hypothalamus, in order to realize the therapeutic effect on true precocious puberty.</p>


Subject(s)
Animals , Female , Rats , Drugs, Chinese Herbal , Pharmacology , Estrus , Gene Expression , Gonadotropin-Releasing Hormone , Genetics , Hypothalamus , Metabolism , Kisspeptins , Genetics , Ovary , RNA, Messenger , Genetics , Metabolism , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled , Genetics , Receptors, Kisspeptin-1 , Reverse Transcriptase Polymerase Chain Reaction , Sexual Maturation , Genetics , Time Factors , Uterus , Vagina
17.
Clinical and Experimental Reproductive Medicine ; : 155-162, 2013.
Article in English | WPRIM | ID: wpr-34817

ABSTRACT

OBJECTIVE: Stress is known to be an inhibitor of the reproductive hypothalamic-pituitary-gonadal (HPG) axis. However, the neural and molecular connections between stress and reproduction are not yet understood. It is well established that in both humans and rodents, kisspeptin (encoded by the kiss1 gene) is a strong stimulator of the HPG axis. In the present study we hypothesized that endocannabinoids, an important neuromodulatory system in the brain, can act on the HPG axis at the level of kiss1 expression to inhibit reproductive function under stress. METHODS: Adult male Wistar rats were unilaterally implanted with an intracerebroventricular cannula. Afterwards, the animals were exposed to immobilization stress, with or without the presence of the cannabinoid CB1 receptor antagonist AM251 (1 microg/rat). Blood samples were collected through a retro-orbital plexus puncture before and after stress. Five hours after the stress, brain tissue was collected for reverse transcriptase-quantitative polymerase chain reaction measurements of kiss1 mRNA. RESULTS: Immobilization stress (1 hour) resulted in a decrease in the serum luteinizing hormone concentration. Additionally, kiss1 gene expression was decreased in key hypothalamic nuclei that regulate gonadotrophin secretion, the medial preoptic area (mPOA), and to some extent the arcuate nucleus (ARC). A single central administration of AM251 was effective in blocking these inhibitory responses. CONCLUSION: These findings suggest that endocannabinoids mediate, at least in part, immobilization stress-induced inhibition of the reproductive system. Our data suggest that the connection between immobilization stress and the HPG axis is kiss1 expression in the mPOA rather than the ARC.


Subject(s)
Adult , Animals , Humans , Male , Rats , Arcuate Nucleus of Hypothalamus , Axis, Cervical Vertebra , Brain , Cannabinoids , Catheters , Endocannabinoids , Gene Expression , Immobilization , Kisspeptins , Luteinizing Hormone , Polymerase Chain Reaction , Preoptic Area , Punctures , Rats, Wistar , Receptor, Cannabinoid, CB1 , Reproduction , RNA, Messenger , Rodentia
18.
Annals of Pediatric Endocrinology & Metabolism ; : 55-59, 2013.
Article in English | WPRIM | ID: wpr-133885

ABSTRACT

Puberty is the end-point of a complex series of developmental events, defined by the dynamic interaction between genetic factors and environmental cues, ultimately leading to the attainment of reproductive capacity. Kisspeptins, products of the KISS1 gene, were originally identified as metastasis suppressor peptides with the ability to bind G protein-coupled receptors (GPR54). In 2003, loss-of-function mutations of the GPR54 gene were found in patients with hypogonadotropic hypogonadism. This finding triggered study of the role of the kisspeptin/GPR54 system as an essential gatekeeper of control of reproduction and pubertal development. Kisspeptins are very potent elicitors of gonadotropin secretion, primarily through stimulation of gonadotropin-releasing hormone release. KISS1 also functions as an essential integrator for peripheral inputs, including gonadal steroids and nutritional signals, and for controlling GnRH and gonadotropin secretion. Whether the kisspeptin/GPR54 system is the trigger for puberty onset and/or it operates as integrator and effector of up-stream regulatory factors warrants further investigation.


Subject(s)
Humans , Cues , Gonadotropin-Releasing Hormone , Gonadotropins , Gonads , Hypogonadism , Kisspeptins , Leptin , Neoplasm Metastasis , Peptides , Puberty , Reproduction , Steroids
19.
Annals of Pediatric Endocrinology & Metabolism ; : 55-59, 2013.
Article in English | WPRIM | ID: wpr-133884

ABSTRACT

Puberty is the end-point of a complex series of developmental events, defined by the dynamic interaction between genetic factors and environmental cues, ultimately leading to the attainment of reproductive capacity. Kisspeptins, products of the KISS1 gene, were originally identified as metastasis suppressor peptides with the ability to bind G protein-coupled receptors (GPR54). In 2003, loss-of-function mutations of the GPR54 gene were found in patients with hypogonadotropic hypogonadism. This finding triggered study of the role of the kisspeptin/GPR54 system as an essential gatekeeper of control of reproduction and pubertal development. Kisspeptins are very potent elicitors of gonadotropin secretion, primarily through stimulation of gonadotropin-releasing hormone release. KISS1 also functions as an essential integrator for peripheral inputs, including gonadal steroids and nutritional signals, and for controlling GnRH and gonadotropin secretion. Whether the kisspeptin/GPR54 system is the trigger for puberty onset and/or it operates as integrator and effector of up-stream regulatory factors warrants further investigation.


Subject(s)
Humans , Cues , Gonadotropin-Releasing Hormone , Gonadotropins , Gonads , Hypogonadism , Kisspeptins , Leptin , Neoplasm Metastasis , Peptides , Puberty , Reproduction , Steroids
20.
Chinese Journal of Hepatology ; (12): 925-929, 2012.
Article in Chinese | WPRIM | ID: wpr-246761

ABSTRACT

<p><b>OBJECTIVE</b>To investigate the impact of expression of kisspeptin-1 (KiSS-1) metastasis-suppressor gene on the proliferative, adhesive and invasive abilities of human hepatocellular carcinoma (HCC) using an in vitro cell system.</p><p><b>METHODS</b>The highly metastatic human hepatoma cell line MHCC97-H was transiently transfected with the pcDNA3.1/HisC vector expressing the KiSS-1 gene (experimental group) or the vector without the KisS-1 gene (blank control group). Untransfected cells served as the negative control group. Proliferative abilities of the three groups were assessed by flow cytometry and MTT assay. Adhesive abilities were assessed by MTT assays using matrigel and fibronectin. Invasive abilities and cell motility were assessed by chemoinvasion chamber assay using reconstituted matrigel and migration chamber assay using polycarbonate filters, respectively.</p><p><b>RESULTS</b>The experimental group showed significantly lower adhesion capacity to matrigel (0.257+/-0.029) than either the blank control group (0.374+/-0.016; t=-7.90345, P less than 0.01) or the negative control group (0.394+/-0.031; t=-7.22752, P less than 0.01). Similarly, the experimental group showed significantly lower adhesion capacity to fibronectin (0.292+/-0.004) than either the blank control group (0.394+/-0.010; t=-20.93138, P less than 0.01) or the negative control group (0.412+/-0.023; t=-11.31371, P less than 0.01). The experimental group also showed significantly lower numbers of cells with invasive capacity (42.40+/-1.14) than either the blank control group (66+/-1.58; t=-27.0711, P less than 0.01) or the negative control group (67.80 +/- 1.92; t=-25.4, P less than 0.01). Similarly, the experimental group showed significantly lower numbers of cells with chemotactic movement (65.80+/-1.92) than either the blank control group (93.80+/-2.28; t=-30.11750, P less than 0.01) or the negative control group (96.40+/-2.07; t=-24.19142, P less than 0.01). The experimental group showed slightly, but not significantly, lower cell proliferation (0.644+/-0.027) than either the blank control group (0.669+/-0.022; t=-1.60371, P?>?0.05) or the negative control group (0.678+/-0.027; t=-1.97828, P?>?0.05). In addition, there were no obvious differences between the three groups in the amounts of cells arrested in either the G1 phase or the S phase.</p><p><b>CONCLUSION</b>KiSS-1 overexpression suppresses the adhesion, invasion and motility, but not the proliferation, of hepatoma carcinoma cells in vitro. These findings imply that KiSS-1 might represent a promising new candidate for gene therapy against human hepatocellular carcinoma.</p>


Subject(s)
Humans , Apoptosis , Carcinoma, Hepatocellular , Pathology , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Proliferation , Kisspeptins , Genetics , Liver Neoplasms , Pathology , Neoplasm Invasiveness , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL