Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Ther Adv Endocrinol Metab ; 14: 20420188231173325, 2023.
Article in English | MEDLINE | ID: mdl-37435528

ABSTRACT

Background: Endometriosis is an oestrogen-dependent disease that can cause subfertility in women who may require assisted reproductive technology (ART) to achieve their pregnancy goals. Objectives: The aim of this study was to compare ART outcomes in women with endometriosis following the long GnRH-agonist controlled ovarian stimulation (COS) protocol with those taking the GnRH-antagonist COS protocol. Data Sources and Methods: MEDLINE, Embase and Web of Science were systematically searched in June 2022. Randomized controlled trials (RCTs) and observational studies comparing the long GnRH-agonist COS protocol and the GnRH-antagonist COS protocol in women with all stages/subtypes of endometriosis were included. Data were synthesized into comprehensive tables for systematic review. The Scottish Intercollegiate Guidelines Network (SIGN) checklists were used for the risk of bias assessment of non-randomized studies and randomized studies, and all the included studies were deemed to have acceptable quality. Main Results: Eight studies (one RCT and seven observational) with 2695 patients (2761 cycles) were included. Most studies generally reported non-significant differences in clinical pregnancy or live birth rates regardless of the COS protocol used. However, the GnRH-agonist protocol may yield a higher total number of oocytes retrieved, especially mature oocytes. Conversely, the GnRH-antagonist protocol required a shorter COS duration and lower gonadotrophin dose. Adverse outcomes, such as rates of cycle cancellation and miscarriage, were similar between both COS protocols. Conclusion: Both the long GnRH-agonist and GnRH-antagonist COS protocols generally yield similar pregnancy outcomes. However, the long GnRH-agonist protocol may be associated with a higher cumulative pregnancy rate due to the higher number of retrieved oocytes available for cryopreservation. The underlying mechanisms of the two COS protocols on the female reproductive tract remain unclear. Clinicians should consider treatment costs, stage/subtype of endometriosis and pregnancy goals of their patients when selecting a GnRH analogue for COS. A well-powered RCT is needed to minimize the risk of bias and compare the risk for ovarian hyperstimulation syndrome. Registration: This review was prospectively registered at PROSPERO under Registration No. CRD42022327604.

2.
Adv Ther ; 38(10): 5025-5045, 2021 10.
Article in English | MEDLINE | ID: mdl-34514552

ABSTRACT

INTRODUCTION: Hot flushes/flashes (HFs) or other vasomotor symptoms affect between 45 and 97% of women during menopause. Hormone replacement therapy (HRT) is effective at alleviating menopausal symptoms, but some women cannot or prefer not to take HRT. Since current non-hormonal options have suboptimal efficacy/tolerability, there is a pressing need for an effective, well-tolerated alternative. The neurokinin 3 receptor (NK3R) has recently been implicated in the generation of menopausal HFs and represents a novel therapeutic target to ameliorate HF symptoms. This review aims to assess if NK3R antagonists (NK3Ras) are more effective than Serotonin Norepinephrine Reuptake Inhibitors (SNRIs)-currently a common choice for non-hormonal treatment of menopausal HFs. METHODS: Studies were identified after systematically searching Ovid MEDLINE and EMBASE databases based on PRISMA guidelines. Trial quality and bias were assessed. Key efficacy outcomes (HF frequency, HF severity and number of night-time awakenings/night-sweats) and selected safety outcomes were extracted and analysed. RESULTS: Seven SNRI and four NK3Ra placebo-controlled randomised trials (plus four follow-up reports) were included in this review. NK3Ra administration resulted in a larger reduction from baseline in HF frequency, HF severity and night-sweats compared to SNRIs. Five of seven SNRI trials showed a reduction in HF frequency that was statistically significant (by 48-67% from baseline at weeks 8 or 12) whereas all NK3Ra trials showed a statistically significant reduction in HF frequency (by 62-93% from baseline at weeks 2, 4 or 12). While SNRI trials reported poor tolerability, particularly nausea, NK3Ra trials reported good tolerability overall, although two trials reported elevation in transaminases. CONCLUSION: NK3Ras trials show encouraging efficacy and tolerability/safety. Completion of phase 3 NK3Ra trials are required to confirm efficacy and uphold safety/tolerability data but phase 2 results suggest that NK3Ras are more effective than SNRIs for non-hormonal treatment of menopausal HFs.


Subject(s)
Selective Serotonin Reuptake Inhibitors , Serotonin , Female , Humans , Menopause , Norepinephrine , Receptors, Neurokinin-3 , Selective Serotonin Reuptake Inhibitors/therapeutic use
3.
J Neuroendocrinol ; 32(2): e12819, 2020 02.
Article in English | MEDLINE | ID: mdl-31800973

ABSTRACT

The present study investigated neuroanatomically localised changes in de novo DNA methyltransferase expression in the female Siberian hamster (Phodopus sungorus). The objectives were to identify the neuroendocrine substrates that exhibit rhythmic Dnmt3a and Dnmt3b expression across the oestrous cycle and also examine the role of ovarian steroids. Hypothalamic Dnmt3a expression was observed to significantly increase during the transition from pro-oestrous to oestrous. A single bolus injection of diethylstilbestrol and progesterone was sufficient to increase Dnmt3a cell numbers and Dnmt3b immunoreactive intensity in the suprachiasmatic nucleus. In vitro analyses using an embryonic rodent cell line revealed that diethylstilbestrol was sufficient to induce Dnmt3b expression. Up-regulating DNA methylation in vitro reduced the expression of vasoactive intestinal polypeptide, Vip, and the circadian clock gene, Bmal1. Together, these data indicate that ovarian steroids drive de novo DNA methyltransferase expression in the mammalian suprachiasmatic nucleus and increased methylation may regulate genes involved in the circadian timing of oestrous: Vip and Bmal1. Overall, epigenetically mediated neuroendocrine reproductive events may reflect an evolutionarily ancient process involved in the timing of female fertility.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , Gonadal Hormones/metabolism , Ovary/metabolism , Suprachiasmatic Nucleus/metabolism , Animals , Circadian Clocks , DNA Methylation , Estrous Cycle/metabolism , Female , Neurosecretory Systems/metabolism , Phodopus
4.
J Comp Neurol ; 527(16): 2634-2643, 2019 11 01.
Article in English | MEDLINE | ID: mdl-30950042

ABSTRACT

Dyslexia is a common neurodevelopmental disorder caused by a significant genetic component. The KIAA0319 gene is one of the most robust dyslexia susceptibility factors but its function remains poorly understood. Initial RNA-interference studies in rats suggested a role in neuronal migration whereas subsequent work with double knock-out mouse models for both Kiaa0319 and its paralogue Kiaa0319-like reported effects in the auditory system but not in neuronal migration. To further understand the role of KIAA0319 during neurodevelopment, we carried out an expression study of its zebrafish orthologue at different embryonic stages. We used different approaches including RNAscope in situ hybridization combined with light-sheet microscopy. The results show particularly high expression during the first few hours of development. Later, expression becomes localized in well-defined structures. In addition to high expression in the brain, we report for the first time expression in the eyes and the notochord. Surprisingly, kiaa0319-like, which generally shows a similar expression pattern to kiaa0319, was not expressed in the notochord suggesting a distinct role for kiaa0319 in this structure. This observation was supported by the identification of notochord enhancers enriched upstream of the KIAA0319 transcription start site, in both zebrafish and humans. This study supports a developmental role for KIAA0319 in the brain as well as in other developing structures, particularly in the notochord which, is key for establishing body patterning in vertebrates.


Subject(s)
Brain/embryology , Brain/metabolism , Eye/embryology , Eye/metabolism , Notochord/metabolism , Animals , Animals, Genetically Modified , Cell Movement/physiology , Dyslexia/genetics , Dyslexia/metabolism , Gene Expression , Gene Expression Regulation, Developmental , Humans , Liver/metabolism , Myocardium/metabolism , Neurogenesis/physiology , Neurons/metabolism , Zebrafish
5.
Adv Biosyst ; 3(3): e1800290, 2019 03.
Article in English | MEDLINE | ID: mdl-32627397

ABSTRACT

Optogenetics, photostimulation of neural tissues rendered sensitive to light, is widely used in neuroscience to modulate the electrical excitability of neurons. For effective optical excitation of neurons, light wavelength and power density must fit with the expression levels and biophysical properties of the genetically encoded light-sensitive ion channels used to confer light sensitivity on cells-most commonly, channelrhodopsins (ChRs). As light sources, organic light-emitting diodes (OLEDs) offer attractive properties for miniaturized implantable devices for in vivo optical stimulation, but they do not yet operate routinely at the optical powers required for optogenetics. Here, OLEDs with doped charge transport layers are demonstrated that deliver blue light with good stability over millions of pulses, at powers sufficient to activate the ChR, CheRiff when expressed in cultured primary neurons, allowing live cell imaging of neural activity with the red genetically encoded calcium indicator, jRCaMP1a. Intracellular calcium responses scale with the radiant flux of OLED emission, when varied through changes in the current density, number of pulses, frequency, and pulse width delivered to the devices. The reported optimization and characterization of high-power OLEDs are foundational for the development of miniaturized OLEDs with thin-layer encapsulation on bioimplantable devices to allow single-cell activation in vivo.


Subject(s)
Neurons , Optogenetics/methods , Photic Stimulation/methods , Animals , Cells, Cultured , Channelrhodopsins/chemistry , Channelrhodopsins/metabolism , Electrodes, Implanted , Hippocampus/cytology , Mice , Neurons/chemistry , Neurons/cytology , Neurons/metabolism , Recombinant Proteins
6.
Neuroendocrinology ; 108(3): 172-189, 2019.
Article in English | MEDLINE | ID: mdl-30537700

ABSTRACT

BACKGROUND/AIMS: The medial amygdala (MeA) responds to olfactory stimuli and alters reproductive physiology. However, the neuronal circuit that relays signals from the MeA to the reproductive axis remains poorly defined. This study aimed to test whether MeA kisspeptin (MeAKiss) neurons in male mice are sensitive to sexually relevant olfactory stimuli and transmit signals to alter reproductive physiology. We also investigated whether MeAKiss neurons have the capacity to elaborate glutamate and GABA neurotransmitters and potentially contribute to reproductive axis regulation. METHODS: Using female urine as a pheromone stimulus, MeAKiss neuronal activity was analysed and serum luteinizing hormone (LH) was measured in male mice. Next, using a chemogenetic approach, MeAKiss neurons were bi-directionally modulated to measure the effect on serum LH and evaluate the activation of the preoptic area. Lastly, using in situ hybridization, we identified the proportion of MeAKiss neurons that express markers for GABAergic (Vgat) and glutamatergic (Vglut2) neurotransmission. RESULTS: Male mice exposed to female urine showed a two-fold increase in the number of c-Fos-positive MeAKiss neurons concomitant with raised LH. Chemogenetic activation of MeAKiss neurons significantly increased LH in the absence of urine exposure, whereas inhibition of MeAKiss neurons did not alter LH. In situ hybridization revealed that MeAKiss neurons are a mixed neuronal population in which 71% express Vgat mRNA, 29% express Vglut2 mRNA, and 6% express both. CONCLUSIONS: Our results uncover, for the first time, that MeAKiss neurons process sexually relevant olfactory signals to influence reproductive hormone levels in male mice, likely through a complex interplay of neuropeptide and neurotransmitter signalling.


Subject(s)
Amygdala/physiology , Kisspeptins/physiology , Luteinizing Hormone/blood , Neurons/physiology , Pheromones/pharmacology , Administration, Inhalation , Amygdala/drug effects , Amygdala/metabolism , Animals , Clozapine/analogs & derivatives , Clozapine/pharmacology , Glutamic Acid/metabolism , Kisspeptins/genetics , Male , Mice , Mice, Transgenic , Pheromones/administration & dosage , Pheromones/urine , Preoptic Area/physiology , Vesicular Glutamate Transport Protein 2/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism , gamma-Aminobutyric Acid/metabolism
7.
Sci Adv ; 4(4): eaar4817, 2018 04.
Article in English | MEDLINE | ID: mdl-29740614

ABSTRACT

Scattering and absorption limit the penetration of optical fields into tissue. We demonstrate a new approach for increased depth penetration in light-sheet microscopy: attenuation-compensation of the light field. This tailors an exponential intensity increase along the illuminating propagation-invariant field, enabling the redistribution of intensity strategically within a sample to maximize signal and minimize irradiation. A key attribute of this method is that only minimal knowledge of the specimen transmission properties is required. We numerically quantify the imaging capabilities of attenuation-compensated Airy and Bessel light sheets, showing that increased depth penetration is gained without compromising any other beam attributes. This powerful yet straightforward concept, combined with the self-healing properties of the propagation-invariant field, improves the contrast-to-noise ratio of light-sheet microscopy up to eightfold across the entire field of view in thick biological specimens. This improvement can significantly increase the imaging capabilities of light-sheet microscopy techniques using Airy, Bessel, and other propagation-invariant beam types, paving the way for widespread uptake by the biomedical community.

8.
Biomed Opt Express ; 8(11): 5179-5190, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-29188112

ABSTRACT

Optical clearing is emerging as a popular approach particularly for studies in neuroscience. However the use of corrosive clearing solutions typically requires sophisticated objectives or extreme care with optical components chosen for single- or multi-photon imaging. In contrast to the use of complex, custom-made microscope objectives, we show that the use of a corrected multimode fibre (MMF) offers a route that is resistant to corrosion, can be used in clearing media, is not constrained by the refractive index of the immersion medium and offers flexible working distances. Using a corrected MMF, we demonstrate fluorescence imaging of beads and stained neuroblastoma cells through optically cleared mouse brain tissue, as well as imaging in an extreme oxidative environment to show the versatility of our approach. Additionally, we perform Raman imaging of polystyrene beads in clearing media to demonstrate that this approach may be used for vibrational spectroscopy of optically cleared samples.

9.
Biomed Opt Express ; 7(10): 4021-4033, 2016 Oct 01.
Article in English | MEDLINE | ID: mdl-27867712

ABSTRACT

We have investigated the effect of Airy illumination on the image quality and depth penetration of digitally scanned light-sheet microscopy in turbid neural tissue. We used Fourier analysis of images acquired using Gaussian and Airy light-sheets to assess their respective image quality versus penetration into the tissue. We observed a three-fold average improvement in image quality at 50 µm depth with the Airy light-sheet. We also used optical clearing to tune the scattering properties of the tissue and found that the improvement when using an Airy light-sheet is greater in the presence of stronger sample-induced aberrations. Finally, we used homogeneous resolution probes in these tissues to quantify absolute depth penetration in cleared samples with each beam type. The Airy light-sheet method extended depth penetration by 30% compared to a Gaussian light-sheet.

10.
Hum Mol Genet ; 25(9): 1771-9, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26908617

ABSTRACT

We recently reported the association of the PCSK6 gene with handedness through a quantitative genome-wide association study (GWAS; P < 0.5 × 10(-8)) for a relative hand skill measure in individuals with dyslexia. PCSK6 activates Nodal, a morphogen involved in regulating left-right body axis determination. Therefore, the GWAS data suggest that the biology underlying the patterning of structural asymmetries may also contribute to behavioural laterality, e.g. handedness. The association is further supported by an independent study reporting a variable number tandem repeat (VNTR) within the same PCSK6 locus to be associated with degree of handedness in a general population cohort. Here, we have conducted a functional analysis of the PCSK6 locus combining further genetic analysis, in silico predictions and molecular assays. We have shown that the previous GWAS signal was not tagging a VNTR effect, suggesting that the two markers have independent effects. We demonstrated experimentally that one of the top GWAS-associated markers, rs11855145, directly alters the binding site for a nuclear factor. Furthermore, we have shown that the predicted regulatory region adjacent to rs11855415 acts as a bidirectional promoter controlling the expression of novel RNA transcripts. These include both an antisense long non-coding RNA (lncRNA) and a short PCSK6 isoform predicted to be coding. This is the first molecular characterization of a handedness-associated locus that supports the role of common variants in non-coding sequences in influencing complex phenotypes through gene expression regulation.


Subject(s)
Functional Laterality/genetics , Gene Expression Regulation , Genome-Wide Association Study , Introns/genetics , Minisatellite Repeats/genetics , Promoter Regions, Genetic/genetics , Proprotein Convertases/genetics , Serine Endopeptidases/genetics , Genetic Variation/genetics , Humans , Nodal Protein/genetics , RNA, Long Noncoding/genetics
11.
Mol Biol Evol ; 31(4): 765-78, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24361996

ABSTRACT

Gonadotropin-releasing hormone (GnRH) is a critical reproductive regulator in vertebrates. Homologous peptides are also found in invertebrates, with a variety of characterized functions. In the amphioxus, an invertebrate that provides the best model for the transition to vertebrates, four GnRH receptors (GnRHRs) were previously described, but their native ligands were not identified. Using a more sensitive search methodology with hidden Markov models, we identified the first GnRH-like peptide confirmed in the amphioxus Branchiostoma floridae. This peptide specifically activated one of the four GnRHRs. Although the primary structure of this peptide was divergent from any previously isolated GnRH peptide, the minimal conserved residues found in all other GnRH superfamily members were retained. The peptide was immunolocalized in proximity of the central canal of the anterior nerve cord, a region where other neuropeptides and receptors have been found. Additionally, the amphioxus GnRH-like gene was positioned in a locus surrounded by syntenic homologs of the human GnRH paralogon. The amphioxus GnRH-like peptide, with its distinct primary structure, activated a receptor with equal potency to multiple ligands that span the GnRH superfamily.


Subject(s)
Gonadotropin-Releasing Hormone/physiology , Lancelets/genetics , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Chlorocebus aethiops , Conserved Sequence , Evolution, Molecular , Gonadotropin-Releasing Hormone/chemistry , Humans , Molecular Sequence Data , Nervous System/metabolism , Organ Specificity , Phylogeny , Receptors, LHRH/metabolism , Signal Transduction , Synteny
12.
Gen Comp Endocrinol ; 190: 134-43, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23756151

ABSTRACT

Diverse external and internal environmental factors are integrated in the hypothalamus to regulate the reproductive system. This is mediated through the pulsatile secretion of GnRH into the portal system to stimulate pituitary gonadotrophin secretion, which in turn regulates gonadal function. A single subpopulation of neurones termed 'KNDy neurones' located in the hypothalamic arcuate nucleus co-localise kisspeptin (Kiss), neurokinin B (NKB) and dynorphin (Dyn) and are responsive to negative feedback effects of sex steroids. The co-ordinated secretion from KNDy neurones appears to modulate the pulsatile release of GnRH, acting as a proximate pacemaker. This review briefly describes the neuropeptidergic control of reproduction in the avian class, highlighting the status of reproductive neuropeptide signalling systems homologous to those found in mammalian genomes. Genes encoding the GnRH system are complete in the chicken with similar roles to the mammalian counterparts, whereas genes encoding Kiss signalling components appear missing in the avian lineage, indicating a differing set of hypothalamic signals controlling avian reproduction. Gene sequences encoding both NKB and Dyn signalling components are present in the chicken genome, but expression analysis and functional studies remain to be completed. The focus of this article is to describe the avian complement of neuropeptidergic reproductive hormones and provide insights into the putative mechanisms that regulate reproduction in birds. These postulations highlight differences in reproductive strategies of birds in terms of gonadal steroid feedback systems, integration of metabolic signals and seasonality. Also included are propositions of KNDy neuropeptide gene silencing and plasticity in utilisation of these neuropeptides during avian evolution.


Subject(s)
Chickens/metabolism , Gonadotropin-Releasing Hormone/metabolism , Neuropeptides/metabolism , Reproduction/physiology , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Dynorphins/metabolism , Kisspeptins/metabolism , Neurokinin B/metabolism
13.
Am J Physiol Endocrinol Metab ; 305(1): E67-77, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23632635

ABSTRACT

The human GnRH receptor (GNRHR1) has a specific set of properties with physiological and pharmacological influences not appropriately modeled in laboratory animals or cell-based systems. To address this deficiency, we have generated human GNRHR1 knock-in mice and described their reproductive phenotype. Measurement of pituitary GNRHR1 transcripts from homozygous human GNRHR1 knock-in (ki/ki) mice revealed a severe reduction (7- to 8-fold) compared with the mouse Gnrhr1 in wild-type mice. ¹²5I-GnRH binding assays on pituitary membrane fractions corroborated reduced human GNRHR1 protein expression in ki/ki mice, as occurs with transfection of human GNRHR1 in cell lines. Female homozygous knock-in mice displayed normal pubertal onset, indicating that a large reduction in GNRHR1 expression is sufficient for this process. However, ki/ki females exhibited periods of prolonged estrous and/or metestrous and reduced fertility. No impairment was found in reproductive maturity or adult fertility in male ki/ki mice. Interestingly, the serum LH response to GnRH challenge was reduced in both knock-in males and females, indicating a reduced GNRHR1 signaling capacity. Small molecules targeting human GPCRs usually have poor activities at homologous rodent receptors, thus limiting their use in preclinical development. Therefore, we tested a human-specific GnRH1 antagonist, NBI-42902, in our mouse model and demonstrated abrogation of a GnRH1-induced serum LH rise in ki/ki mice and an absence of effect in littermates expressing the wild-type murine receptor. This novel model provides the opportunity to study the human receptor in vivo and for screening the activity of human-specific GnRH analogs.


Subject(s)
Estrous Cycle/physiology , Fertility/physiology , Receptors, LHRH/genetics , Receptors, LHRH/physiology , Reproduction/physiology , Animals , Feedback, Physiological/drug effects , Feedback, Physiological/physiology , Female , Gene Knock-In Techniques , Humans , Male , Mice , Mice, Transgenic , Models, Animal , Phenotype , Pituitary Gland/physiology , Pregnancy , Receptors, LHRH/antagonists & inhibitors , Sexual Maturation/physiology , Testis/growth & development , Testis/physiology , Thymine/analogs & derivatives , Thymine/pharmacology
14.
Neuroendocrinology ; 97(2): 193-202, 2013.
Article in English | MEDLINE | ID: mdl-22377698

ABSTRACT

Pulsatile gonadotropin-releasing hormone (GnRH) is crucial to normal reproductive function and abnormalities in pulse frequency give rise to reproductive dysfunction. Kisspeptin and neurokinin B (NKB), neuropeptides secreted by the same neuronal population in the ventral hypothalamus, have emerged recently as critical central regulators of GnRH and thus gonadotropin secretion. Patients with mutations resulting in loss of signaling by either of these neuroendocrine peptides fail to advance through puberty but the mechanisms mediating this remain unresolved. We report here that continuous kisspeptin infusion restores gonadotropin pulsatility in patients with loss-of-function mutations in NKB (TAC3) or its receptor (TAC3R), indicating that kisspeptin on its own is sufficient to stimulate pulsatile GnRH secretion. Moreover, our findings suggest that NKB action is proximal to kisspeptin in the reproductive neuroendocrine cascade regulating GnRH secretion, and may act as an autocrine modulator of kisspeptin secretion. The ability of continuous kisspeptin infusion to induce pulsatile gonadotropin secretion further indicates that GnRH neurons are able to set up pulsatile secretion in the absence of pulsatile exogenous kisspeptin.


Subject(s)
Disorders of Sex Development/genetics , Kisspeptins/administration & dosage , Luteinizing Hormone/metabolism , Neurokinin B/deficiency , Receptors, Neurokinin-3/genetics , Adult , Disorders of Sex Development/blood , Disorders of Sex Development/physiopathology , Disorders of Sex Development/therapy , Estradiol/blood , Female , Humans , Inhibins/blood , Luteinizing Hormone/blood , Male , Mutation/physiology , Neurokinin B/genetics , Pulsatile Flow/drug effects , Receptors, Neurokinin-3/deficiency , Receptors, Neurokinin-3/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Testosterone/blood
15.
PLoS One ; 7(6): e38456, 2012.
Article in English | MEDLINE | ID: mdl-22679506

ABSTRACT

Congenital hypogonadotropic hypogonadism (CHH) is characterized by low gonadotropins and failure to progress normally through puberty. Mutations in the gene encoding the GnRH receptor (GNRHR1) result in CHH when present as compound heterozygous or homozygous inactivating mutations. This study identifies and characterizes the properties of two novel GNRHR1 mutations in a family in which three brothers display normosmic CHH while their sister was unaffected. Molecular analysis in the proband and the affected brothers revealed two novel non-synonymous missense GNRHR1 mutations, present in a compound heterozygous state, whereas their unaffected parents possessed only one inactivating mutation, demonstrating the autosomal recessive transmission in this kindred and excluding X-linked inheritance equivocally suggested by the initial pedigree analysis. The first mutation at c.845 C>G introduces an Arg substitution for the conserved Pro 282 in transmembrane domain (TMD) 6. The Pro282Arg mutant is unable to bind radiolabeled GnRH analogue. As this conserved residue is important in receptor conformation, it is likely that the mutation perturbs the binding pocket and affects trafficking to the cell surface. The second mutation at c.968 A>G introduces a Cys substitution for Tyr 323 in the functionally crucial N/DPxxY motif in TMD 7. The Tyr323Cys mutant has an increased GnRH binding affinity but reduced receptor expression at the plasma membrane and impaired G protein-coupling. Inositol phosphate accumulation assays demonstrated absent and impaired Gα(q/11) signal transduction by Pro282Arg and Tyr323Cys mutants, respectively. Pretreatment with the membrane permeant GnRHR antagonist NBI-42902, which rescues cell surface expression of many GNRHR1 mutants, significantly increased the levels of radioligand binding and intracellular signaling of the Tyr323Cys mutant but not Pro282Arg. Immunocytochemistry confirmed that both mutants are present on the cell membrane albeit at low levels. Together these molecular deficiencies of the two novel GNRHR1 mutations lead to the CHH phenotype when present as a compound heterozygote.


Subject(s)
Hypogonadism/genetics , Receptors, LHRH/genetics , Adolescent , Adult , Animals , COS Cells , Cell Membrane/metabolism , Chlorocebus aethiops , Female , Heterozygote , Humans , Hypogonadism/congenital , Hypogonadism/metabolism , Inositol Phosphates/metabolism , Male , Middle Aged , Mutation/genetics , Young Adult
16.
N Engl J Med ; 366(7): 629-35, 2012 Feb 16.
Article in English | MEDLINE | ID: mdl-22335740

ABSTRACT

Gonadotropin-releasing hormone (GnRH) is the central regulator of gonadotropins, which stimulate gonadal function. Hypothalamic neurons that produce kisspeptin and neurokinin B stimulate GnRH release. Inactivating mutations in the genes encoding the human kisspeptin receptor (KISS1R, formerly called GPR54), neurokinin B (TAC3), and the neurokinin B receptor (TACR3) result in pubertal failure. However, human kisspeptin loss-of-function mutations have not been described, and contradictory findings have been reported in Kiss1-knockout mice. We describe an inactivating mutation in KISS1 in a large consanguineous family that results in failure of pubertal progression, indicating that functional kisspeptin is important for puberty and reproduction in humans. (Funded by the Scientific and Technological Research Council of Turkey [TÜBITAK] and others.).


Subject(s)
Hypogonadism/genetics , Kisspeptins/genetics , Mutation , Puberty/genetics , Adolescent , Adult , Child , Consanguinity , Female , Genes, Recessive , Genotyping Techniques , Gonadotropin-Releasing Hormone/metabolism , Humans , Male , Pedigree , Sequence Analysis, DNA
17.
Brain Res ; 1364: 81-9, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-20858467

ABSTRACT

Kisspeptin has recently been identified as a key neuroendocrine gatekeeper of reproduction and is essential for the initiation of human puberty and maintenance of adult reproduction. Kisspeptin neurons appear to be integrative sensors, as they respond to changes in numerous internal and external factors including nutrient and fat status, stress and sex steroids, thus providing a link between these factors and reproduction. We have pioneered the development of kisspeptin antagonists as powerful tools for interrogating the role of kisspeptin in reproductive physiology and pathology, and as potential treatments for hormone-dependent disease. This article summarizes their development and key findings to date. These demonstrate an essential role for kisspeptin in GnRH neuron firing, GnRH pulsatile secretion, negative feedback by gonadal steroids, the onset of puberty, and the ovulatory LH surge. These studies establish that kisspeptin antagonists are powerful investigative tools and set the scene for more extensive physiological and pathophysiological studies as well as therapeutic intervention.


Subject(s)
Reproduction/physiology , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/physiology , Animals , Blood-Brain Barrier/physiology , Electrophysiological Phenomena/drug effects , Electrophysiological Phenomena/physiology , Feedback, Physiological/drug effects , Feedback, Physiological/physiology , Female , Gonadotropin-Releasing Hormone/physiology , Humans , Kisspeptins , Male , Neurons/physiology , Puberty/physiology
18.
N Engl J Med ; 360(26): 2742-8, 2009 Jun 25.
Article in English | MEDLINE | ID: mdl-19535795

ABSTRACT

We investigated whether mutations in the gene encoding gonadotropin-releasing hormone 1 (GNRH1) might be responsible for idiopathic hypogonadotropic hypogonadism (IHH) in humans. We identified a homozygous GNRH1 frameshift mutation, an insertion of an adenine at nucleotide position 18 (c.18-19insA), in the sequence encoding the N-terminal region of the signal peptide-containing protein precursor of gonadotropin-releasing hormone (prepro-GnRH) in a teenage brother and sister, who had normosmic IHH. Their unaffected parents and a sibling who was tested were heterozygous. This mutation results in an aberrant peptide lacking the conserved GnRH decapeptide sequence, as shown by the absence of immunoreactive GnRH when expressed in vitro. This isolated autosomal recessive GnRH deficiency, reversed by pulsatile GnRH administration, shows the pivotal role of GnRH in human reproduction.


Subject(s)
Frameshift Mutation , Gonadotropin-Releasing Hormone/genetics , Hypogonadism/genetics , Protein Precursors/genetics , Adolescent , Estradiol/blood , Female , Follicle Stimulating Hormone/blood , Genes, Recessive , Gonadotropin-Releasing Hormone/administration & dosage , Gonadotropin-Releasing Hormone/deficiency , Homozygote , Humans , Luteinizing Hormone/blood , Luteinizing Hormone/metabolism , Male , Pedigree , Sequence Analysis, DNA , Testosterone/blood
19.
Endocrinology ; 150(6): 2847-56, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19264870

ABSTRACT

In vertebrates, activation of the GnRH receptor is necessary to initiate the reproductive cascade. However, little is known about the characteristics of GnRH receptors before the vertebrates evolved. Recently genome sequencing was completed for amphioxus, Branchiostoma floridae. To understand the GnRH receptors (GnRHR) from this most basal chordate, which is also classified as an invertebrate, we cloned and characterized four GnRHR cDNAs encoded in the amphioxus genome. We found that incubation of GnRH1 (mammalian GnRH) and GnRH2 (chicken GnRH II) with COS7 cells heterologously expressing the amphioxus GnRHRs caused potent intracellular inositol phosphate turnover in two of the receptors. One of the two receptors displayed a clear preference for GnRH1 over GnRH2, a characteristic not previously seen outside the type I mammalian GnRHRs. Phylogenetic analysis grouped the four receptors into two paralogous pairs, with one pair grouping basally with the vertebrate GnRH receptors and the other grouping with the octopus GnRHR-like sequence and the related receptor for insect adipokinetic hormone. Pharmacological studies showed that octopus GnRH-like peptide and adipokinetic hormone induced potent inositol phosphate turnover in one of these other two amphioxus receptors. These data demonstrate the functional conservation of two distinct types of GnRH receptors at the base of chordates. We propose that one receptor type led to vertebrate GnRHRs, whereas the other type, related to the mollusk GnRHR-like receptor, was lost in the vertebrate lineage. This is the first report to suggest that distinct invertebrate and vertebrate GnRHRs are present simultaneously in a basal chordate, amphioxus.


Subject(s)
Evolution, Molecular , Invertebrates , Phylogeny , Receptors, LHRH/analysis , Receptors, LHRH/genetics , Vertebrates , Amino Acid Sequence , Animals , Chordata , DNA, Complementary/genetics , Humans , Molecular Sequence Data , Receptors, LHRH/physiology , Reproduction/physiology , Signal Transduction/physiology
20.
Gen Comp Endocrinol ; 156(2): 201-9, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18178211

ABSTRACT

Successful reproduction in vertebrates depends upon the actions of gonadotropin-releasing hormone (GnRH). Despite the wide presence of GnRH in Phylum Chordata, GnRH has not been isolated in protostomes other than the common octopus. To provide information on the evolution of this critical hormone, we isolated the full-length cDNA of a GnRH-like molecule from the central nervous system of a gastropod mollusk, the sea hare Aplysia californica. The open reading frame of this cDNA encodes a protein of 147 amino acids. The molecular architecture of the deduced protein is highly homologous to that reported for the prepro-octopus GnRH (oct-GnRH) and consists of a putative signal peptide, a GnRH dodecapeptide, a downstream processing site, and a GnRH-associated peptide (GAP). The deduced amino acid sequence of the Aplysia GnRH (ap-GnRH) is QNYHFSNGWYAG and differs from oct-GnRH by only two amino acids. The transcript for ap-GnRH is widely expressed in the central nervous system (CNS), the ovotestis, and the atrial gland, an exocrine gland. Immunocytochemistry (ICC) using an antiserum against oct-GnRH detected immunoreactive neurons in all CNS ganglia examined, and the staining was abolished by the preadsorption of the antiserum with synthetic ap-GnRH. In sum, ap-GnRH sequence is the first gastropod GnRH-like molecule to be elucidated. Further, it represents one of the only two GnRH-like molecules found outside Phylum Chordata. These data refute the possibility that oct-GnRH arose singly in cephalopods by convergent evolution and provide valuable support for an ancient origin of GnRH during metazoan evolution.


Subject(s)
Aplysia/physiology , Gonadotropin-Releasing Hormone/metabolism , Amino Acid Sequence , Animals , Aplysia/genetics , Cloning, Molecular , DNA Primers , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Databases, Factual , Gene Expression Regulation/genetics , Immunohistochemistry , Molecular Sequence Data , Neuropeptides/physiology , Phylogeny , RNA/biosynthesis , RNA/genetics , RNA/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...