Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Am J Obstet Gynecol ; 208(2): 132.e1-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23174285

ABSTRACT

OBJECTIVE: We evaluated vaginal defensin concentrations and levels of bacterial vaginosis-associated bacterial species in pregnant women. STUDY DESIGN: Self-collected vaginal swabs from 2 visits during pregnancy were tested with quantitative polymerase chain reaction for 9 bacterial species. Beta defensins 2-3 and alpha defensins 1-3 were measured by enzyme-linked immunosorbent assay. RESULTS: Our 126 participants were primarily African American (60%), had a mean gestational age at enrollment of 10 ± 3 weeks and at follow-up visit of 25 ± 6 weeks. At enrollment, the prevalence of bacterial vaginosis was 74% (94/126 women), which decreased to 60% (75/126 specimens) at follow-up visit. At enrollment, beta defensin 3 concentrations were significantly lower in women with bacterial vaginosis (2.64 ± 0.91 vs 3.25 ± 0.99 log(10) pg/mL; P = .003). Higher concentrations of Atopobium vaginae, bacterial vaginosis-associated bacteria1 and 2 were associated with significantly lower concentrations of beta defensin 3 (P < .01). CONCLUSION: Bacterial vaginosis was associated with lower vaginal concentrations of beta defensin 3, but not beta defensin 2 or alpha defensins 1-3, in pregnant women.


Subject(s)
Bacteria/isolation & purification , Pregnancy Complications, Infectious , Vaginosis, Bacterial/microbiology , alpha-Defensins/metabolism , beta-Defensins/metabolism , Adolescent , Adult , Body Fluids/metabolism , Cohort Studies , Enzyme-Linked Immunosorbent Assay , Female , Follow-Up Studies , Gestational Age , Humans , Polymerase Chain Reaction , Pregnancy , Prevalence , Prospective Studies , Vagina/metabolism , Vaginal Smears , Vaginosis, Bacterial/metabolism , Young Adult
2.
Endocrinology ; 152(11): 4298-309, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21933870

ABSTRACT

Neurons that produce kisspeptin play a critical role in reproduction. However, understanding the molecular physiology of kisspeptin neurons has been limited by the lack of an in vivo marker for those cells. Here, we report the development of a Kiss1-CreGFP knockin mouse, wherein the endogenous Kiss1 promoter directs the expression of a Cre recombinase-enhanced green fluorescent protein (GFP) fusion protein. The pattern of GFP expression in the brain of the knockin recapitulates what has been described earlier for Kiss1 in the male and female mouse, with prominent expression in the arcuate nucleus (ARC) (in both sexes) and the anteroventral periventricular nucleus (in females). Single-cell RT-PCR showed that the Kiss1 transcript is expressed in 100% of GFP-labeled cells, and the CreGFP transcript was regulated by estradiol in the same manner as the Kiss1 gene (i.e. inhibited in the ARC and induced in the anteroventral periventricular nucleus). We used this mouse to evaluate the biophysical properties of kisspeptin (Kiss1) neurons in the ARC of the female mouse. GFP-expressing Kiss1 neurons were identified in hypothalamic slice preparations of the ARC and patch clamped. Whole-cell (and loose attached) recordings revealed that Kiss1 neurons exhibit spontaneous activity and expressed both h- (pacemaker) and T-type calcium currents, and hyperpolarization-activated cyclic nucleotide-regulated 1-4 and CaV3.1 channel subtypes (measured by single cell RT-PCR), respectively. N-methyl-D-aspartate induced bursting activity, characterized by depolarizing/hyperpolarizing oscillations. Therefore, Kiss1 neurons in the ARC share molecular and electrophysiological properties of other CNS pacemaker neurons.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Kisspeptins/metabolism , Neurons/metabolism , Reproduction/physiology , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Estradiol/pharmacology , Female , Male , Mice , Mice, Transgenic , Neurons/drug effects , Orchiectomy , Ovariectomy , Reproduction/drug effects
3.
J Neurosci ; 29(38): 11859-66, 2009 Sep 23.
Article in English | MEDLINE | ID: mdl-19776272

ABSTRACT

Kisspeptin is encoded by the Kiss1 gene, and kisspeptin signaling plays a critical role in reproduction. In rodents, kisspeptin neurons in the arcuate nucleus (Arc) provide tonic drive to gonadotropin-releasing hormone (GnRH) neurons, which in turn supports basal luteinizing hormone (LH) secretion. Our objectives were to determine whether preprodynorphin (Dyn) and neurokinin B (NKB) are coexpressed in Kiss1 neurons in the mouse and to evaluate its physiological significance. Using in situ hybridization, we found that Kiss1 neurons in the Arc of female mice not only express the Dyn and NKB genes but also the NKB receptor gene (NK3) and the Dyn receptor [the kappa opioid receptor (KOR)] gene. We also found that expression of the Dyn, NKB, KOR, and NK3 in the Arc are inhibited by estradiol, as has been established for Kiss1, and confirmed that Dyn and NKB inhibit LH secretion. Moreover, using Dyn and KOR knock-out mice, we found that long-term disruption of Dyn/KOR signaling compromises the rise of LH after ovariectomy. We propose a model whereby NKB and dynorphin act autosynaptically on kisspeptin neurons in the Arc to synchronize and shape the pulsatile secretion of kisspeptin and drive the release of GnRH from fibers in the median eminence.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Dynorphins/metabolism , Gonadotropin-Releasing Hormone/metabolism , Neurokinin B/metabolism , Neurons/metabolism , Protein Precursors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Dynorphins/genetics , Estradiol/metabolism , Female , In Situ Hybridization , Kisspeptins , Luteinizing Hormone/blood , Luteinizing Hormone/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Neurological , Ovariectomy , Protein Precursors/genetics , RNA, Messenger/metabolism , Receptors, Neurokinin-3/agonists , Receptors, Neurokinin-3/metabolism , Receptors, Opioid/agonists , Receptors, Opioid/genetics , Receptors, Opioid/metabolism , Signal Transduction
4.
J Neurosci ; 29(29): 9390-5, 2009 Jul 22.
Article in English | MEDLINE | ID: mdl-19625529

ABSTRACT

Kisspeptin is a product of the Kiss1 gene and is expressed in the forebrain. Neurons that express Kiss1 play a crucial role in the regulation of pituitary luteinizing hormone secretion and reproduction. These neurons are the direct targets for the action of estradiol-17beta (E(2)), which acts via the estrogen receptor alpha isoform (ER alpha) to regulate Kiss1 expression. In the arcuate nucleus (Arc), where the dynorphin gene (Dyn) is expressed in Kiss1 neurons, E(2) inhibits the expression of Kiss1 mRNA. However, E(2) induces the expression of Kiss1 in the anteroventral periventricular nucleus (AVPV). The mechanism for differential regulation of Kiss1 in the Arc and AVPV by E(2) is unknown. ER alpha signals through multiple pathways, which can be categorized as either classical, involving the estrogen response element (ERE), or nonclassical, involving ERE-independent mechanisms. To elucidate the molecular basis for the action of E(2) on Kiss1 and Dyn expression, we studied the effects of E(2) on Kiss1 and Dyn mRNAs in the brains of mice bearing targeted alterations in the ER alpha signaling pathways. We found that stimulation of Kiss1 expression by E(2) in the AVPV and inhibition of Dyn in the Arc required an ERE-dependent pathway, whereas the inhibition of Kiss1 expression by E(2) in the Arc involved ERE-independent mechanisms. Thus, distinct ER alpha signaling pathways can differentially regulate the expression of identical genes across different brain regions, and E(2) can act within the same neuron through divergent ER alpha signaling pathways to regulate different neurotransmitter genes.


Subject(s)
Brain/drug effects , Dynorphins/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Estrogens/pharmacology , Proteins/metabolism , Animals , Anterior Thalamic Nuclei/drug effects , Anterior Thalamic Nuclei/metabolism , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Brain/metabolism , Dynorphins/genetics , Female , Gene Expression Regulation/drug effects , Gene Knock-In Techniques , Kisspeptins , Luteinizing Hormone/blood , Mice , Mice, Transgenic , Midline Thalamic Nuclei/drug effects , Midline Thalamic Nuclei/metabolism , Neurons/drug effects , Neurons/metabolism , Proteins/genetics , RNA, Messenger/metabolism , Signal Transduction
5.
J Neurosci ; 29(12): 3920-9, 2009 Mar 25.
Article in English | MEDLINE | ID: mdl-19321788

ABSTRACT

Neurons that produce gonadotropin-releasing hormone (GnRH) are the final common pathway by which the brain regulates reproduction. GnRH neurons are regulated by an afferent network of kisspeptin-producing neurons. Kisspeptin binds to its cognate receptor on GnRH neurons and stimulates their activity, which in turn provides an obligatory signal for GnRH secretion, thus gating down-stream events supporting reproduction. We have developed kisspeptin antagonists to facilitate the direct determination of the role of kisspeptin neurons in the neuroendocrine regulation of reproduction. In vitro and in vivo studies of analogues of kisspeptin-10 with amino substitutions have identified several potent and specific antagonists. A selected antagonist was shown to inhibit the firing of GnRH neurons in the brain of the mouse and to reduce pulsatile GnRH secretion in female pubertal monkeys; the later supporting a key role of kisspeptin in puberty onset. This analog also inhibited the kisspeptin-induced release of luteinizing hormone (LH) in rats and mice and blocked the postcastration rise in LH in sheep, rats, and mice, suggesting that kisspeptin neurons mediate the negative feedback effect of sex steroids on gonadotropin secretion in mammals. The development of kisspeptin antagonists provides a valuable tool for investigating the physiological and pathophysiological roles of kisspeptin in the regulation of reproduction and could offer a unique therapeutic agent for treating hormone-dependent disorders of reproduction, including precocious puberty, endometriosis, and metastatic prostate cancer.


Subject(s)
Gonadotropin-Releasing Hormone/physiology , Peptides/pharmacology , Tumor Suppressor Proteins/antagonists & inhibitors , Action Potentials , Animals , Brain/physiology , CHO Cells , Castration , Cricetinae , Cricetulus , Female , Gonadotropin-Releasing Hormone/metabolism , Humans , In Vitro Techniques , Kisspeptins , Luteinizing Hormone/metabolism , Macaca mulatta , Male , Mice , Microdialysis , Peptides/chemistry , Rats , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/genetics , Receptors, Kisspeptin-1 , Sheep , Structure-Activity Relationship , Tumor Suppressor Proteins/chemistry
6.
Peptides ; 30(1): 4-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18644415

ABSTRACT

The cancer suppressor gene, KISS1, was initially described as having an important role in inhibiting cancer metastasis. Since then, KISS1 and its receptor, KISS1R, have been shown to play a key role in controlling the onset of puberty of reproductive physiology in the human and other species. Recent studies have also linked KISS1/kisspeptin/KISS1R to other processes, such as vasoconstriction, aging, adipocyte physiology, and perhaps as a molecular conduit linking metabolism and reproduction. This article highlights the history of KISS1/kisspeptin/KISS1R biology and proposes a consensus for nomenclature of the key molecules in this signaling pathway.


Subject(s)
Receptors, G-Protein-Coupled , Terminology as Topic , Tumor Suppressor Proteins , Animals , Humans , Hypothalamus/cytology , Hypothalamus/metabolism , Kisspeptins , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, Kisspeptin-1 , Reproduction/physiology , Signal Transduction/physiology , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
7.
J Neurosci ; 27(44): 12088-95, 2007 Oct 31.
Article in English | MEDLINE | ID: mdl-17978050

ABSTRACT

The Kiss1 gene codes for kisspeptin, which binds to GPR54, a G-protein-coupled receptor. Kisspeptin and GPR54 are expressed in discrete regions of the forebrain, and they have been implicated in the neuroendocrine regulation of reproduction. Kiss1-expressing neurons are thought to regulate the secretion of gonadotropin-releasing hormone (GnRH) and thus coordinate the estrous cycle in rodents; however, the precise role of kisspeptin-GPR54 signaling in the regulation of gonadotropin secretion is unknown. In this study, we used female mice with deletions in the GPR54 gene [GPR54 knock-outs (KOs)] to test the hypothesis that kisspeptin-GPR54 signaling provides the drive necessary for tonic GnRH/luteinizing hormone (LH) release. We predicted that tonic GnRH/LH secretion would be disrupted in GPR54 KOs and that such animals would be incapable of showing a compensatory rise in LH secretion after ovariectomy. As predicted, we found that GPR54 KO mice do not exhibit a postovariectomy rise in LH, suggesting that tonic GnRH secretion is disrupted in the absence of kisspeptin-GPR54 signaling. We also postulated that kisspeptin-GPR54 signaling is critical for the generation of the estradiol (E)-induced GnRH/LH surge and thus E should be incapable of inducing an LH surge in the absence of GPR54. However, we found that E induced Fos expression in GnRH neurons and produced a GnRH-dependent LH surge in GPR54 KOs. Thus, in mice, kisspeptin-GPR54 signaling is required for the tonic stimulation of GnRH/LH secretion but is not required for generating the E-induced GnRH/LH surge.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Proteins/metabolism , Receptors, G-Protein-Coupled/physiology , Signal Transduction/physiology , Animals , Behavior, Animal , Brain/cytology , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation/genetics , Kisspeptins , Luteinizing Hormone/blood , Mice , Mice, Knockout , Neurons/metabolism , Oligopeptides/pharmacology , Oncogene Proteins v-fos/genetics , Oncogene Proteins v-fos/metabolism , Ovariectomy/methods , Proteins/genetics , Radioimmunoassay/methods , Receptors, G-Protein-Coupled/deficiency , Receptors, Kisspeptin-1
8.
J Neurosci ; 27(33): 8826-35, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17699664

ABSTRACT

GPR54 is a G-protein-coupled receptor, which binds kisspeptins and is widely expressed throughout the brain. Kisspeptin-GPR54 signaling has been implicated in the regulation of pubertal and adulthood gonadotropin-releasing hormone (GnRH) secretion, and mutations or deletions of GPR54 cause hypogonadotropic hypogonadism in humans and mice. Other reproductive roles for kisspeptin-GPR54 signaling, including the regulation of developmental GnRH secretion or sexual behavior in adults, have not yet been explored. Using adult wild-type (WT) and GPR54 knock-out (KO) mice, we first tested whether kisspeptin-GPR54 signaling is necessary for male and female sexual behaviors. We found that hormone-replaced gonadectomized GPR54 KO males and females displayed appropriate gender-specific adult sexual behaviors. Next, we examined whether GPR54 signaling is required for proper display of olfactory-mediated partner preference behavior. Testosterone-treated WT males preferred stimulus females rather than males, whereas similarly treated WT females and GPR54 KO males showed no preference for either sex. Because olfactory preference is sexually dimorphic and organized during development by androgens, we assessed whether GPR54 signaling is essential for sexual differentiation of other sexually dimorphic traits. Interestingly, adult testosterone-treated GPR54 KO males displayed "female-like" numbers of tyrosine hydroxylase-immunoreactive and Kiss1 mRNA-containing neurons in the anteroventral periventricular nucleus and likewise possessed fewer motoneurons in the spino-bulbocavernosus nucleus than did WT males. Our findings indicate that kisspeptin-GPR54 signaling is not required for male or female copulatory behavior, provided there is appropriate adulthood hormone replacement. However, GPR54 is necessary for proper male-like development of several sexually dimorphic traits, likely by regulating GnRH-mediated androgen secretion during "critical windows" in perinatal development.


Subject(s)
Brain/metabolism , Receptors, G-Protein-Coupled/physiology , Sex Differentiation/physiology , Sexual Behavior, Animal/physiology , Signal Transduction/physiology , Analysis of Variance , Animals , Behavior, Animal/physiology , Brain/cytology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Gonadotropin-Releasing Hormone/metabolism , Humans , Kisspeptins , Luteinizing Hormone/blood , Male , Mice , Mice, Knockout , Neurons/classification , Neurons/drug effects , Neurons/metabolism , Proteins/genetics , Proteins/metabolism , Receptors, G-Protein-Coupled/deficiency , Receptors, Kisspeptin-1 , Sex Characteristics , Sex Differentiation/drug effects , Sex Differentiation/genetics , Sexual Behavior, Animal/drug effects , Signal Transduction/drug effects , Testosterone/pharmacology , Tumor Suppressor Proteins/pharmacology , Tyrosine 3-Monooxygenase/metabolism
9.
Endocrinology ; 148(4): 1774-83, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17204549

ABSTRACT

The Kiss1 gene codes for kisspeptins, which have been implicated in the neuroendocrine regulation of reproduction. In the brain, Kiss1 mRNA-expressing neurons are located in the arcuate (ARC) and anteroventral periventricular (AVPV) nuclei. Kiss1 neurons in the AVPV appear to play a role in generating the preovulatory GnRH/LH surge, which occurs only in females and is organized perinatally by gonadal steroids. Because Kiss1 is involved in the sexually dimorphic GnRH/LH surge, we hypothesized that Kiss1 expression is sexually differentiated, with females having more Kiss1 neurons than either males or neonatally androgenized females. To test this, male and female rats were neonatally treated with androgen or vehicle; then, as adults, they were left intact or gonadectomized and implanted with capsules containing sex steroids or nothing. Kiss1 mRNA levels in the AVPV and ARC were determined by in situ hybridization. Normal females expressed significantly more Kiss1 mRNA in the AVPV than normal males, even under identical adult hormonal conditions. This Kiss1 sex difference was organized perinatally, as demonstrated by the observation that neonatally androgenized females displayed a male-like pattern of adulthood Kiss1 expression in the AVPV. In contrast, there was neither a sex difference nor an influence of neonatal treatment on Kiss1 expression in the ARC. Using double-labeling techniques, we determined that the sexually differentiated Kiss1 neurons in the AVPV are distinct from the sexually differentiated population of tyrosine hydroxylase (dopaminergic) neurons in this region. Our findings suggest that sex differences in kisspeptin signaling from the AVPV subserve the cellular mechanisms controlling the sexually differentiated GnRH/LH surge.


Subject(s)
Brain/metabolism , Proteins/genetics , Sex Characteristics , Sex Differentiation/genetics , Animals , Animals, Newborn , Female , Gene Expression Regulation, Developmental , Gonadotropins/blood , Kisspeptins , Male , Proteins/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar
10.
J Neurosci ; 25(49): 11349-56, 2005 Dec 07.
Article in English | MEDLINE | ID: mdl-16339030

ABSTRACT

We examined the role of kisspeptin and its receptor, the G-protein-coupled receptor GPR54, in governing the onset of puberty in the mouse. In the adult male and female mouse, kisspeptin (10-100 nM) evoked a remarkably potent, long-lasting depolarization of >90% of gonadotropin-releasing hormone (GnRH)-green fluorescent protein neurons in situ. In contrast, in juvenile [postnatal day 8 (P8) to P19] and prepubertal (P26-P33) male mice, kisspeptin activated only 27 and 44% of GnRH neurons, respectively. This developmental recruitment of GnRH neurons into a kisspeptin-responsive pool was paralleled by an increase in the ability of centrally administered kisspeptin to evoke luteinizing hormone secretion in vivo. To learn more about the mechanisms through which kisspeptin-GPR54 signaling at the GnRH neuron may change over postnatal development, we performed quantitative in situ hybridization for kisspeptin and GPR54 transcripts. Approximately 90% of GnRH neurons were found to express GPR54 mRNA in both juvenile and adult mice, without a detectable difference in the mRNA content between the age groups. In contrast, the expression of KiSS-1 mRNA increased dramatically across the transition from juvenile to adult life in the anteroventral periventricular nucleus (AVPV; p < 0.001). These results demonstrate that kisspeptin exerts a potent depolarizing effect on the excitability of almost all adult GnRH neurons and that the responsiveness of GnRH neurons to kisspeptin increases over postnatal development. Together, these observations suggest that activation of GnRH neurons by kisspeptin at puberty reflects a dual process involving an increase in kisspeptin input from the AVPV and a post-transcriptional change in GPR54 signaling within the GnRH neuron.


Subject(s)
Action Potentials/physiology , Gonadotropin-Releasing Hormone/metabolism , Neurons/metabolism , Neurosecretory Systems/physiology , Proteins/physiology , Sexual Maturation/physiology , Age Factors , Animals , Animals, Newborn , Anterior Thalamic Nuclei/metabolism , Anterior Thalamic Nuclei/physiology , Female , Kisspeptins , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Mutant Strains , Neurons/physiology , Neurosecretory Systems/growth & development , Proteins/pharmacology , Tumor Suppressor Proteins
11.
Mol Cell Biol ; 25(11): 4804-11, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15899880

ABSTRACT

Galanin is a neuropeptide implicated in the regulation of feeding, reproduction, cognition, nociception, and seizure susceptibility. There are three known galanin receptor (GALR) subtypes (GALR1, GALR2, and GALR3), which bind to galanin with different affinities and have their own unique distributions, signaling mechanisms, and putative functions in the brain and peripheral nervous system. To gain further insight into the possible physiological significance of GALR2, we created mutant mice that were deficient in GALR2 and compared their phenotype to that of wild-type (WT) littermate or age-matched controls, with respect to basic motor and sensory function, feeding behavior, reproduction, mood, learning and memory, and seizure susceptibility. Phenotypic analysis revealed that animals bearing a deletion of GALR2 did not differ significantly from their WT controls in any of the measured variables. We conclude that either GALR2 plays no role in these physiological functions or through redundancy or compensation these mutant animals can adapt to the congenital absence of GALR2. It is also conceivable that GALR2 plays only a subtle role in some of these functions and that the impact of its loss could not be detected by the analytical procedures used here.


Subject(s)
Phenotype , Receptor, Galanin, Type 2/physiology , Animals , Body Weight/genetics , Feeding Behavior , Female , Gene Deletion , Learning , Male , Memory , Mice , Mice, Knockout , Receptor, Galanin, Type 2/deficiency , Receptor, Galanin, Type 2/genetics , Reproduction/genetics , Seizures/genetics , Sex Factors
12.
Endocrinology ; 146(7): 2976-84, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15831567

ABSTRACT

Kisspeptins are products of the Kiss1 gene, which bind to GPR54, a G protein-coupled receptor. Kisspeptins and GPR54 have been implicated in the neuroendocrine regulation of GnRH secretion. To test the hypothesis that testosterone regulates Kiss1 gene expression, we compared the expression of KiSS-1 mRNA among groups of intact, castrated, and castrated/testosterone (T)-treated male mice. In the arcuate nucleus (Arc), castration resulted in a significant increase in KiSS-1 mRNA, which was completely reversed with T replacement, whereas in the anteroventral periventricular nucleus, the results were the opposite, i.e. castration decreased and T increased KiSS-1 mRNA expression. In the Arc, the effects of T on KiSS-1 mRNA were completely mimicked by estrogen but only partially mimicked by dihydrotestosterone, a nonaromatizable androgen, suggesting that both estrogen receptor (ER) and androgen receptor (AR) play a role in T-mediated regulation of KiSS-1. Studies of the effects of T on KiSS-1 expression in mice with either a deletion of the ERalpha or a hypomorphic allele to the AR revealed that the effects of T are mediated by both ERalpha and AR pathways, which was confirmed by the presence of either ERalpha or AR coexpression in most KiSS-1 neurons in the Arc. These observations suggest that KiSS-1 neurons in the Arc, whose transcriptional activity is inhibited by T, are targets for the negative feedback regulation of GnRH secretion, whereas KiSS-1 neurons in the anteroventral periventricular nucleus, whose activity is stimulated by T, may mediate other T-dependent processes.


Subject(s)
Brain/metabolism , Proteins/genetics , RNA, Messenger/metabolism , Testosterone/physiology , Animals , Body Weight , Dihydrotestosterone/pharmacology , Estradiol/pharmacology , Estrogen Receptor alpha/deficiency , Estrogen Receptor alpha/genetics , Hormones/blood , In Situ Hybridization , Kisspeptins , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orchiectomy , Prosencephalon/drug effects , Prosencephalon/metabolism , Receptors, Androgen/deficiency , Receptors, Androgen/genetics , Testosterone/pharmacology , Tissue Distribution
13.
Trends Endocrinol Metab ; 15(5): 215-21, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15223051

ABSTRACT

The arcuate nucleus is a hypothalamic center that couples energetics and reproduction. Peptide-releasing neurons in the arcuate nucleus receive and process humoral signals from the periphery and relay this information to other nuclei in the hypothalamus and preoptic area. Galanin-like peptide (GALP) is expressed in the arcuate nucleus, and GALP-containing neurons are targets for the action of leptin. GALP-containing neurons are closely apposed to gonadotropin-releasing hormone (GnRH) neurons in the preoptic area, and CNS injections of GALP stimulate GnRH-mediated secretion of luteinizing hormone. These observations indicate that GALP is a molecular signal that couples circulating indices of metabolism to the neuroendocrine reproductive system and, thus, regulates reproductive activity as a function of the energy state. In this article, we describe the involvement of GALP in metabolism and reproduction, and in the coupling between these two processes.


Subject(s)
Energy Metabolism/physiology , Galanin-Like Peptide/metabolism , Reproduction/physiology , Animals , Humans
14.
Neuroendocrinology ; 80(4): 264-72, 2004.
Article in English | MEDLINE | ID: mdl-15665556

ABSTRACT

The KiSS-1 gene codes for a family of neuropeptides called kisspeptins which bind to the G-protein-coupled receptor GPR54. To assess the possible effects of kisspeptins on gonadotropin secretion, we injected kisspeptin-52 into the lateral cerebral ventricles of adult male rats and found that kisspeptin-52 increased the serum levels of luteinizing hormone (p < 0.05). To determine whether the kisspeptin-52-induced stimulation of luteinizing hormone secretion was mediated by gonadotropin-releasing hormone (GnRH), we pretreated adult male rats with a GnRH antagonist (acyline), then challenged the animals with intracerebroventricularly administered kisspeptin-52. The GnRH antagonist blocked the kisspeptin-52-induced increase in luteinizing hormone. To examine whether kisspeptins stimulate transcriptional activity in GnRH neurons, we administered kisspeptin-52 intracerebroventricularly and found by immunocytochemistry that 86% of the GnRH neurons coexpressed Fos 2 h after the kisspeptin-52 challenge, whereas fewer than 1% of the GnRH neurons expressed Fos following injection of the vehicle alone (p < 0.001). To assess whether kisspeptins can directly act on GnRH neurons, we used double-label in situ hybridization and found that 77% of the GnRH neurons coexpress GPR54 mRNA. Finally, to determine whether KiSS-1 gene expression is regulated by gonadal hormones, we measured KiSS-1 mRNA levels by single-label in situ hybridization in intact and castrated males and found significantly higher levels in the arcuate nucleus of castrates. These results demonstrate that GnRH neurons are direct targets for regulation by kisspeptins and that KiSS-1 mRNA is regulated by gonadal hormones, suggesting that KiSS-1 neurons play an important role in the feedback regulation of gonadotropin secretion.


Subject(s)
Gene Expression Regulation/drug effects , Gonadotropin-Releasing Hormone/metabolism , Neurons/drug effects , Proteins/metabolism , RNA, Messenger/metabolism , Animals , Castration/methods , Cell Count/methods , Drug Interactions , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Immunohistochemistry/methods , In Situ Hybridization/methods , Injections, Intraventricular/methods , Kisspeptins , Luteinizing Hormone/blood , Male , Neurons/metabolism , Oligopeptides/pharmacology , Oncogene Proteins v-fos/genetics , Oncogene Proteins v-fos/metabolism , Peptides/pharmacology , Proteins/genetics , Radioimmunoassay/methods , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled , Receptors, Kisspeptin-1 , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Statistics, Nonparametric
15.
Connect Tissue Res ; 44(1): 50-7, 2003.
Article in English | MEDLINE | ID: mdl-12945804

ABSTRACT

The connective tissue matrix of the wall of ovarian follicles is degraded and remodeled during ovulatory rupture and formation of the corpus luteum. Ovarian surface epithelial cells in close contact with the apical wall of preovulatory ovine follicles secrete a urokinase-type plasminogen activator in response to surge levels of gonadotropins. Urokinase activates latent collagenases and stimulates release of tumor necrosis factor alpha from thecal endothelium. Tumor necrosis factor alpha progressively induces matrix metalloproteinase gene expression, apoptosis, and inflammatory necrosis. Collagenolysis and cellular death are a prelude to stigma formation and ovarian rupture. Ovulation is blocked by intrafollicular injection of TNFalpha or MMP-2 antisera. Unruptured follicles luteinize but are deficient in collagenous/vascularized trabeculae, and produce less progesterone than their control luteal counterparts. It appears that TNFalpha, via MMP-2 induction, contributes to the reorganization of an ovulatory follicle into a fully competent corpus luteum.


Subject(s)
Luteinization/physiology , Matrix Metalloproteinases/metabolism , Ovarian Follicle/enzymology , Sheep/metabolism , Animals , Female , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...