Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Elife ; 102021 07 22.
Article in English | MEDLINE | ID: mdl-34292152

ABSTRACT

GnRH neurons are the final central neural output regulating fertility. Kisspeptin neurons in the hypothalamic arcuate nucleus (KNDy neurons) are considered the main regulator of GnRH output. GnRH and KNDy neurons are surrounded by astrocytes, which can modulate neuronal activity and communicate over distances. Prostaglandin E2 (PGE2), synthesized primarily by astrocytes, increases GnRH neuron activity and downstream pituitary release of luteinizing hormone (LH). We hypothesized that glial fibrillary acidic protein (GFAP)-expressing astrocytes play a role in regulating GnRH and/or KNDy neuron activity and LH release. We used adeno-associated viruses to target designer receptors exclusively activated by designer drugs (DREADDs) to GFAP-expressing cells to activate Gq- or Gi-mediated signaling. Activating Gq signaling in the preoptic area, near GnRH neurons, but not in the arcuate, increases LH release in vivo and GnRH firing in vitro via a mechanism in part dependent upon PGE2. These data suggest that astrocytes can activate GnRH/LH release in a manner independent of KNDy neurons.


Subject(s)
Arcuate Nucleus of Hypothalamus/drug effects , Glial Fibrillary Acidic Protein/metabolism , Gonadotropin-Releasing Hormone/pharmacology , Luteinizing Hormone/drug effects , Neurons/metabolism , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/metabolism , Kisspeptins/metabolism , Luteinizing Hormone/metabolism , Male , Mice , Mice, Transgenic , Pituitary Gland/metabolism , Preoptic Area/metabolism
2.
Med Sci (Paris) ; 37(4): 366-371, 2021 Apr.
Article in French | MEDLINE | ID: mdl-33908854

ABSTRACT

The survival of the species depends on two closely interlinked processes: the correct functioning of the reproductive system, and the balance between the energy needs of an individual and the supply of energy sources through feeding. These two processes are regulated in the hypothalamus, which produces neurohormones that control various physiological functions. Among these neurohormones, GnRH controls not only the maturation and function of the reproductive organs, including the ovaries and the testes, during puberty and in adulthood, but also sexual attraction. Recent evidence suggest that neuropilin-1-mediated signaling in GnRH-synthesizing neurons could be a linchpin that holds together various neuroanatomical, physiological and behavioral adaptations involved in triggering puberty and achieving reproductive function.


TITLE: Signalisation impliquant la neuropiline dans les neurones sécrétant la GnRH - Son rôle dans le déclenchement de la puberté. ABSTRACT: La survie d'une espèce dépend de deux processus intimement liés : la reproduction, d'une part, et l'équilibre entre les besoins énergétiques et l'approvisionnement en sources d'énergie par l'alimentation, d'autre part. Ces deux processus sont contrôlés dans le cerveau par l'hypothalamus, qui produit des neurohormones agissant sur l'hypophyse pour piloter diverses fonctions physiologiques. L'une de ces neurohormones, la GnRH, contrôle non seulement la maturation et le fonctionnement des organes reproducteurs, incluant les ovaires et les testicules, lors de la puberté et à l'âge adulte, mais aussi l'attirance sexuelle. De récentes découvertes suggèrent que la signalisation impliquant la neuropiline-1 dans les neurones sécrétant la GnRH jouerait un rôle charnière dans la coordination du neurodéveloppement et des adaptations physiologiques et comportementales nécessaires au déclenchement de la puberté et à l'acquisition de la fonction de reproduction. Dans cet article de synthèse, nous replaçons ces découvertes dans le contexte de récents travaux montrant que les voies de signalisation des sémaphorines de classe 3 sont impliquées dans la physiopathologie non seulement de l'infertilité, mais aussi de l'obésité. Nous discutons également l'implication potentielle des neurones produisant la GnRH dans la perception des odeurs sociales et dans la précocité de la maturation sexuelle. L'hypothèse selon laquelle l'activité de ces neurones au cours du développement postnatal constituerait le chaînon manquant entre la prise de poids, le déclenchement de la puberté et le comportement sexuel, ouvre la voie à une meilleure compréhension de l'implication de l'homéostasie énergétique dans la maturation sexuelle, et pourrait aussi avoir des implications thérapeutiques pour la puberté précoce.


Subject(s)
Gonadotropin-Releasing Hormone/biosynthesis , Neurons/metabolism , Neuropilin-1/metabolism , Puberty, Precocious/etiology , Puberty/physiology , Animals , Energy Intake , Energy Metabolism/physiology , Female , Genitalia/physiology , Humans , Hypothalamus/physiology , Male , Mice , Reproduction/physiology , Sex Characteristics , Sexual Arousal
3.
EMBO J ; 39(19): e104633, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32761635

ABSTRACT

Hypothalamic neurons expressing gonadotropin-releasing hormone (GnRH), the "master molecule" regulating reproduction and fertility, migrate from their birthplace in the nose to their destination using a system of guidance cues, which include the semaphorins and their receptors, the neuropilins and plexins, among others. Here, we show that selectively deleting neuropilin-1 in new GnRH neurons enhances their survival and migration, resulting in excess neurons in the hypothalamus and in their unusual accumulation in the accessory olfactory bulb, as well as an acceleration of mature patterns of activity. In female mice, these alterations result in early prepubertal weight gain, premature attraction to male odors, and precocious puberty. Our findings suggest that rather than being influenced by peripheral energy state, GnRH neurons themselves, through neuropilin-semaphorin signaling, might engineer the timing of puberty by regulating peripheral adiposity and behavioral switches, thus acting as a bridge between the reproductive and metabolic axes.


Subject(s)
Gene Expression Regulation , Gonadotropin-Releasing Hormone/metabolism , Neurons/metabolism , Neuropilin-1/biosynthesis , Sexual Behavior, Animal , Sexual Maturation , Weight Gain , Animals , Female , Gonadotropin-Releasing Hormone/genetics , Male , Mice , Mice, Transgenic , Neuropilin-1/genetics
4.
Elife ; 82019 04 04.
Article in English | MEDLINE | ID: mdl-30946012

ABSTRACT

The brain regulates fertility through gonadotropin-releasing hormone (GnRH) neurons. Estradiol induces negative feedback on pulsatile GnRH/luteinizing hormone (LH) release and positive feedback generating preovulatory GnRH/LH surges. Negative and positive feedbacks are postulated to be mediated by kisspeptin neurons in arcuate and anteroventral periventricular (AVPV) nuclei, respectively. Kisspeptin-specific ERα knockout mice exhibit disrupted LH pulses and surges. This knockout approach is neither location-specific nor temporally controlled. We utilized CRISPR-Cas9 to disrupt ERα in adulthood. Mice with ERα disruption in AVPV kisspeptin neurons have typical reproductive cycles but blunted LH surges, associated with decreased excitability of these neurons. Mice with ERα knocked down in arcuate kisspeptin neurons showed disrupted cyclicity, associated with increased glutamatergic transmission to these neurons. These observations suggest that activational effects of estradiol regulate surge generation and maintain cyclicity through AVPV and arcuate kisspeptin neurons, respectively, independent from its role in the development of hypothalamic kisspeptin neurons or puberty onset.


Subject(s)
Hypothalamus/physiology , Neurons/physiology , Reproduction , Sexual Behavior, Animal , Animals , Estradiol/metabolism , Estrogen Receptor alpha/deficiency , Female , Gene Knockout Techniques , Kisspeptins/analysis , Mice, Knockout , Neurons/chemistry
5.
Endocrinology ; 159(4): 1922-1940, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29522155

ABSTRACT

Gonadotropin-releasing hormone (GnRH) neurons are a nexus of fertility regulation. We used translating ribosome affinity purification coupled with RNA sequencing to examine messenger RNAs of GnRH neurons in adult intact and gonadectomized (GDX) male and female mice. GnRH neuron ribosomes were tagged with green fluorescent protein (GFP) and GFP-labeled polysomes isolated by immunoprecipitation, producing one RNA fraction enhanced for GnRH neuron transcripts and one RNA fraction depleted. Complementary DNA libraries were created from each fraction and 50-base, paired-end sequencing done and differential expression (enhanced fraction/depleted fraction) determined with a threshold of >1.5- or <0.66-fold (false discovery rate P ≤ 0.05). A core of ∼840 genes was differentially expressed in GnRH neurons in all treatments, including enrichment for Gnrh1 (∼40-fold), and genes critical for GnRH neuron and/or gonadotrope development. In contrast, non-neuronal transcripts were not enriched or were de-enriched. Several epithelial markers were also enriched, consistent with the olfactory epithelial origins of GnRH neurons. Interestingly, many synaptic transmission pathways were de-enriched, in accordance with relatively low innervation of GnRH neurons. The most striking difference between intact and GDX mice of both sexes was a marked downregulation of genes associated with oxidative phosphorylation and upregulation of glucose transporters in GnRH neurons from GDX mice. This may suggest that GnRH neurons switch to an alternate fuel to increase adenosine triphosphate production in the absence of negative feedback when GnRH release is elevated. Knowledge of the GnRH neuron translatome and its regulation can guide functional studies and can be extended to disease states, such as polycystic ovary syndrome.


Subject(s)
Gene Expression , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Neurons/metabolism , Animals , Female , Green Fluorescent Proteins/metabolism , Male , Mice , Sequence Analysis, RNA
6.
Endocrinology ; 158(10): 3553-3564, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28938398

ABSTRACT

Pulsatile release of gonadotropin-releasing hormone (GnRH) is key to fertility. Pulse frequency is modulated by gonadal steroids and likely arises subsequent to coordination of GnRH neuron firing activity. The source of rhythm generation and the site of steroid feedback remain critical unanswered questions. Arcuate neurons that synthesize kisspeptin, neurokinin B, and dynorphin (KNDy) may be involved in both of these processes. We tested the hypotheses that action potential firing in KNDy neurons is episodic and that gonadal steroids regulate this pattern. Targeted extracellular recordings were made of green fluorescent protein-identified KNDy neurons in brain slices from adult male mice that were intact, castrated, or castrated and treated with estradiol or dihydrotestosterone (DHT). KNDy neurons exhibited marked peaks and nadirs in action potential firing activity during recordings lasting 1 to 3.5 hours. Peaks, identified by Cluster analysis, occurred more frequently in castrated than intact mice, and either estradiol or DHT in vivo or blocking neurokinin type 3 receptor in vitro restored peak frequency to intact levels. The frequency of peaks in firing rate and estradiol regulation of this frequency is similar to that observed for GnRH neurons, whereas DHT suppressed firing in KNDy but not GnRH neurons. We further examined the patterning of action potentials to identify bursts that may be associated with increased neuromodulator release. Burst frequency and duration are increased in castrated compared with intact and steroid-treated mice. The observation that KNDy neurons fire in an episodic manner that is regulated by steroid feedback is consistent with a role for these neurons in GnRH pulse generation and regulation.


Subject(s)
Action Potentials/drug effects , Androgens/pharmacology , Arcuate Nucleus of Hypothalamus/cytology , Dihydrotestosterone/pharmacology , Estradiol/pharmacology , Estrogens/pharmacology , Neurons/drug effects , Orchiectomy , Action Potentials/physiology , Animals , Brain/drug effects , Brain/metabolism , Brain/physiology , Cluster Analysis , Dynorphins/metabolism , Gonadotropin-Releasing Hormone/metabolism , Green Fluorescent Proteins/genetics , Kisspeptins/genetics , Kisspeptins/metabolism , Male , Mice , Mice, Transgenic , Neurokinin B/metabolism , Neurons/metabolism , Neurons/physiology , Patch-Clamp Techniques , Receptors, Neurokinin-3/antagonists & inhibitors
7.
Diabetologia ; 60(7): 1314-1324, 2017 07.
Article in English | MEDLINE | ID: mdl-28456865

ABSTRACT

AIMS/HYPOTHESIS: Regulation of energy balance involves the participation of many factors, including nutrients, among which are circulating lipids, acting as peripheral signals informing the central nervous system of the energy status of the organism. It has been shown that neuronal lipoprotein lipase (LPL) participates in the control of energy balance by hydrolysing lipid particles enriched in triacylglycerols. Here, we tested the hypothesis that LPL in the mediobasal hypothalamus (MBH), a well-known nucleus implicated in the regulation of metabolic homeostasis, could also contribute to the regulation of body weight and glucose homeostasis. METHODS: We injected an adeno-associated virus (AAV) expressing Cre-green fluorescent protein into the MBH of Lpl-floxed mice (and wild-type mice) to specifically decrease LPL activity in the MBH. In parallel, we injected an AAV overexpressing Lpl into the MBH of wild-type mice. We then studied energy homeostasis and hypothalamic ceramide content. RESULTS: The partial deletion of Lpl in the MBH in mice led to an increase in body weight compared with controls (37.72 ± 0.7 g vs 28.46 ± 0.12, p < 0.001) associated with a decrease in locomotor activity. These mice developed hyperinsulinaemia and glucose intolerance. This phenotype also displayed reduced expression of Cers1 in the hypothalamus as well as decreased concentration of several C18 species of ceramides and a 3-fold decrease in total ceramide intensity. Conversely, overexpression of Lpl specifically in the MBH induced a decrease in body weight. CONCLUSIONS/INTERPRETATION: Our study shows that LPL in the MBH is an important regulator of body weight and glucose homeostasis.


Subject(s)
Glucose/metabolism , Hypothalamus/metabolism , Lipoprotein Lipase/metabolism , Weight Gain , Animals , Body Composition , Body Weight , Calorimetry , Ceramides/metabolism , Dependovirus , Gene Deletion , Glucose Tolerance Test , Green Fluorescent Proteins/metabolism , Homeostasis , Hydrolysis , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Time Factors , Triglycerides/blood
8.
Hum Mol Genet ; 25(15): 3208-3215, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27288456

ABSTRACT

Prader-Willi syndrome (PWS) is a genetic disorder characterized by a variety of physiological and behavioral dysregulations, including hyperphagia, a condition that can lead to life-threatening obesity. Feeding behavior is a highly complex process with multiple feedback loops that involve both peripheral and central systems. The arcuate nucleus of the hypothalamus (ARH) is critical for the regulation of homeostatic processes including feeding, and this nucleus develops during neonatal life under of the influence of both environmental and genetic factors. Although much attention has focused on the metabolic and behavioral outcomes of PWS, an understanding of its effects on the development of hypothalamic circuits remains elusive. Here, we show that mice lacking Magel2, one of the genes responsible for the etiology of PWS, display an abnormal development of ARH axonal projections. Notably, the density of anorexigenic α-melanocyte-stimulating hormone axons was reduced in adult Magel2-null mice, while the density of orexigenic agouti-related peptide fibers in the mutant mice appeared identical to that in control mice. On the basis of previous findings showing a pivotal role for metabolic hormones in hypothalamic development, we also measured leptin and ghrelin levels in Magel2-null and control neonates and found that mutant mice have normal leptin and ghrelin levels. In vitro experiments show that Magel2 directly promotes axon growth. Together, these findings suggest that a loss of Magel2 leads to the disruption of hypothalamic feeding circuits, an effect that appears to be independent of the neurodevelopmental effects of leptin and ghrelin and likely involves a direct neurotrophic effect of Magel2.


Subject(s)
Antigens, Neoplasm/metabolism , Ghrelin/metabolism , Hypothalamus/embryology , Leptin/metabolism , Proteins/metabolism , Animals , Antigens, Neoplasm/genetics , Ghrelin/genetics , Leptin/genetics , Mice , Mice, Mutant Strains , Prader-Willi Syndrome/embryology , Prader-Willi Syndrome/genetics , Proteins/genetics
9.
J Clin Invest ; 124(6): 2550-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24812663

ABSTRACT

The transition to puberty and adult fertility both require a minimum level of energy availability. The adipocyte-derived hormone leptin signals the long-term status of peripheral energy stores and serves as a key metabolic messenger to the neuroendocrine reproductive axis. Humans and mice lacking leptin or its receptor fail to complete puberty and are infertile. Restoration of leptin levels in these individuals promotes sexual maturation, which requires the pulsatile, coordinated delivery of gonadotropin-releasing hormone to the pituitary and the resulting surge of luteinizing hormone (LH); however, the neural circuits that control the leptin-mediated induction of the reproductive axis are not fully understood. Here, we found that leptin coordinated fertility by acting on neurons in the preoptic region of the hypothalamus and inducing the synthesis of the freely diffusible volume-based transmitter NO, through the activation of neuronal NO synthase (nNOS) in these neurons. The deletion of the gene encoding nNOS or its pharmacological inhibition in the preoptic region blunted the stimulatory action of exogenous leptin on LH secretion and prevented the restoration of fertility in leptin-deficient female mice by leptin treatment. Together, these data indicate that leptin plays a central role in regulating the hypothalamo-pituitary-gonadal axis in vivo through the activation of nNOS in neurons of the preoptic region.


Subject(s)
Leptin/metabolism , Nitric Oxide/metabolism , Preoptic Area/physiology , Reproduction/physiology , Animals , Female , Humans , Kisspeptins/metabolism , Leptin/deficiency , Leptin/genetics , Luteinizing Hormone/metabolism , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Nitric Oxide Synthase Type I/deficiency , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/metabolism , Preoptic Area/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Kisspeptin-1 , Receptors, Leptin/deficiency , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Signal Transduction
10.
PLoS Biol ; 12(3): e1001808, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24618750

ABSTRACT

Neuropilin-1 (Nrp1) guides the development of the nervous and vascular systems, but its role in the mature brain remains to be explored. Here we report that the expression of the 65 kDa isoform of Sema3A, the ligand of Nrp1, by adult vascular endothelial cells, is regulated during the ovarian cycle and promotes axonal sprouting in hypothalamic neurons secreting gonadotropin-releasing hormone (GnRH), the neuropeptide controlling reproduction. Both the inhibition of Sema3A/Nrp1 signaling and the conditional deletion of Nrp1 in GnRH neurons counteract Sema3A-induced axonal sprouting. Furthermore, the localized intracerebral infusion of Nrp1- or Sema3A-neutralizing antibodies in vivo disrupts the ovarian cycle. Finally, the selective neutralization of endothelial-cell Sema3A signaling in adult Sema3aloxP/loxP mice by the intravenous injection of the recombinant TAT-Cre protein alters the amplitude of the preovulatory luteinizing hormone surge, likely by perturbing GnRH release into the hypothalamo-hypophyseal portal system. Our results identify a previously unknown function for 65 kDa Sema3A-Nrp1 signaling in the induction of axonal growth, and raise the possibility that endothelial cells actively participate in synaptic plasticity in specific functional domains of the adult central nervous system, thus controlling key physiological functions such as reproduction.


Subject(s)
Brain/metabolism , Endothelial Cells/metabolism , Fertility/physiology , Neuropilin-1/physiology , Semaphorin-3A/metabolism , Animals , Axons/metabolism , Axons/ultrastructure , Estrous Cycle/metabolism , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/physiology , Ligands , Luteinizing Hormone/metabolism , Mice , Mice, Inbred C57BL , Neuropilin-1/metabolism , Rats , Rats, Sprague-Dawley , Semaphorin-3A/genetics , Semaphorin-3A/physiology , Signal Transduction
11.
Endocrinology ; 155(5): 1887-98, 2014 May.
Article in English | MEDLINE | ID: mdl-24601879

ABSTRACT

Traumatic brain injury is a leading cause of hypopituitarism, which compromises patients' recovery, quality of life, and life span. To date, there are no means other than standardized animal studies to provide insights into the mechanisms of posttraumatic hypopituitarism. We have found that GH levels were impaired after inducing a controlled cortical impact (CCI) in mice. Furthermore, GHRH stimulation enhanced GH to lower level in injured than in control or sham mice. Because many characteristics were unchanged in the pituitary glands of CCI mice, we looked for changes at the hypothalamic level. Hypertrophied astrocytes were seen both within the arcuate nucleus and the median eminence, two pivotal structures of the GH axis, spatially remote to the injury site. In the arcuate nucleus, GHRH neurons were unaltered. In the median eminence, injured mice exhibited unexpected alterations. First, the distributions of claudin-1 and zonula occludens-1 between tanycytes were disorganized, suggesting tight junction disruptions. Second, endogenous IgG was increased in the vicinity of the third ventricle, suggesting abnormal barrier properties after CCI. Third, intracerebroventricular injection of a fluorescent-dextran derivative highly stained the hypothalamic parenchyma only after CCI, demonstrating an increased permeability of the third ventricle edges. This alteration of the third ventricle might jeopardize the communication between the hypothalamus and the pituitary gland. In conclusion, the phenotype of CCI mice had similarities to the posttraumatic hypopituitarism seen in humans with intact pituitary gland and pituitary stalk. It is the first report of a pathological status in which tanycyte dysfunctions appear as a major acquired syndrome.


Subject(s)
Brain Injuries/physiopathology , Disease Models, Animal , Ependymoglial Cells/pathology , Hypopituitarism/etiology , Hypothalamus/pathology , Neurons/pathology , Tight Junctions/pathology , Animals , Arcuate Nucleus of Hypothalamus/immunology , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/pathology , Biomarkers/metabolism , Ependymoglial Cells/immunology , Ependymoglial Cells/metabolism , Gene Expression Regulation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Growth Hormone-Releasing Hormone/genetics , Growth Hormone-Releasing Hormone/metabolism , Hypopituitarism/immunology , Hypopituitarism/metabolism , Hypopituitarism/pathology , Hypothalamus/immunology , Hypothalamus/metabolism , Immunoglobulin G/metabolism , Male , Median Eminence/immunology , Median Eminence/metabolism , Median Eminence/pathology , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/immunology , Neurons/metabolism , Permeability , Recombinant Fusion Proteins/metabolism , Third Ventricle/immunology , Third Ventricle/metabolism , Third Ventricle/pathology , Tight Junctions/immunology , Tight Junctions/metabolism
12.
PLoS Genet ; 8(8): e1002896, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22927827

ABSTRACT

Kallmann syndrome (KS) associates congenital hypogonadism due to gonadotropin-releasing hormone (GnRH) deficiency and anosmia. The genetics of KS involves various modes of transmission, including oligogenic inheritance. Here, we report that Nrp1(sema/sema) mutant mice that lack a functional semaphorin-binding domain in neuropilin-1, an obligatory coreceptor of semaphorin-3A, have a KS-like phenotype. Pathohistological analysis of these mice indeed showed abnormal development of the peripheral olfactory system and defective embryonic migration of the neuroendocrine GnRH cells to the basal forebrain, which results in increased mortality of newborn mice and reduced fertility in adults. We thus screened 386 KS patients for the presence of mutations in SEMA3A (by Sanger sequencing of all 17 coding exons and flanking splice sites) and identified nonsynonymous mutations in 24 patients, specifically, a frameshifting small deletion (D538fsX31) and seven different missense mutations (R66W, N153S, I400V, V435I, T688A, R730Q, R733H). All the mutations were found in heterozygous state. Seven mutations resulted in impaired secretion of semaphorin-3A by transfected COS-7 cells (D538fsX31, R66W, V435I) or reduced signaling activity of the secreted protein in the GN11 cell line derived from embryonic GnRH cells (N153S, I400V, T688A, R733H), which strongly suggests that these mutations have a pathogenic effect. Notably, mutations in other KS genes had already been identified, in heterozygous state, in five of these patients. Our findings indicate that semaphorin-3A signaling insufficiency contributes to the pathogenesis of KS and further substantiate the oligogenic pattern of inheritance in this developmental disorder.


Subject(s)
Axons/metabolism , Kallmann Syndrome/genetics , Mutation , Neuropilin-1/metabolism , Semaphorin-3A/genetics , Animals , Disease Models, Animal , Embryo, Mammalian/metabolism , Female , Fetus/metabolism , Gonadotropin-Releasing Hormone/metabolism , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Neuropilin-1/genetics , Nose/innervation , Semaphorin-3A/chemistry , Semaphorin-3A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...