Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters

Complementary Medicines
Database
Language
Affiliation country
Publication year range
1.
JCI Insight ; 8(8)2023 04 24.
Article in English | MEDLINE | ID: mdl-37092553

ABSTRACT

Makorin ring finger protein 3 (MKRN3) was identified as an inhibitor of puberty initiation with the report of loss-of-function mutations in association with central precocious puberty. Consistent with this inhibitory role, a prepubertal decrease in Mkrn3 expression was observed in the mouse hypothalamus. Here, we investigated the mechanisms of action of MKRN3 in the central regulation of puberty onset. We showed that MKRN3 deletion in hypothalamic neurons derived from human induced pluripotent stem cells was associated with significant changes in expression of genes controlling hypothalamic development and plasticity. Mkrn3 deletion in a mouse model led to early puberty onset in female mice. We found that Mkrn3 deletion increased the number of dendritic spines in the arcuate nucleus but did not alter the morphology of GnRH neurons during postnatal development. In addition, we identified neurokinin B (NKB) as an Mkrn3 target. Using proteomics, we identified insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1) as another target of MKRN3. Interactome analysis revealed that IGF2BP1 interacted with MKRN3, along with several members of the polyadenylate-binding protein family. Our data show that one of the mechanisms by which MKRN3 inhibits pubertal initiation is through regulation of prepubertal hypothalamic development and plasticity, as well as through effects on NKB and IGF2BP1.


Subject(s)
Induced Pluripotent Stem Cells , Puberty, Precocious , Humans , Female , Mice , Animals , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Induced Pluripotent Stem Cells/metabolism , Hypothalamus/metabolism , Puberty , Gonadotropin-Releasing Hormone/metabolism , Puberty, Precocious/genetics , Puberty, Precocious/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
2.
Endocrinology ; 163(11)2022 10 11.
Article in English | MEDLINE | ID: mdl-35974456

ABSTRACT

Makorin ring finger protein 3 (MKRN3) is an important neuroendocrine player in the control of pubertal timing and upstream inhibitor of gonadotropin-releasing hormone secretion. In mice, expression of Mkrn3 in the hypothalamic arcuate and anteroventral periventricular nucleus is high early in life and declines before the onset of puberty. Therefore, we aimed to explore if the persistence of hypothalamic Mkrn3 expression peripubertally would result in delayed puberty. Female mice that received neonatal bilateral intracerebroventricular injections of a recombinant adeno-associated virus expressing Mkrn3 had delayed vaginal opening and first estrus compared with animals injected with control virus. Subsequent estrous cycles and fertility were normal. Interestingly, male mice treated similarly did not exhibit delayed puberty onset. Kiss1, Tac2, and Pdyn mRNA levels were increased in the mediobasal hypothalamus in females at postnatal day 28, whereas kisspeptin and neurokinin B protein levels in the arcuate nucleus were decreased, following Mkrn3 overexpression, compared to controls. Cumulatively, these data suggest that Mkrn3 may directly or indirectly target neuropeptides of Kiss1 neurons to degradation pathways. This mouse model suggests that MKRN3 may be a potential contributor to delayed onset of puberty, in addition to its well-established roles in central precocious puberty and the timing of menarche.


Subject(s)
Hypothalamus , Sexual Maturation , Ubiquitin-Protein Ligases , Animals , Female , Gonadotropin-Releasing Hormone , Hypothalamus/metabolism , Kisspeptins/genetics , Male , Mice , Neurokinin B/genetics , Sexual Maturation/genetics , Ubiquitin-Protein Ligases/genetics
3.
J Neurosci ; 35(37): 12903-16, 2015 Sep 16.
Article in English | MEDLINE | ID: mdl-26377475

ABSTRACT

The gonadotropin-releasing hormone (GnRH) is the master regulator of fertility and kisspeptin (KP) is a potent trigger of GnRH secretion from GnRH neurons. KP signals via KISS1R, a Gαq/11-coupled receptor, and mice bearing a global deletion of Kiss1r (Kiss1r(-/-)) or a GnRH neuron-specific deletion of Kiss1r (Kiss1r(d/d)) display hypogonadotropic hypogonadism and infertility. KISS1R also signals via ß-arrestin, and in mice lacking ß-arrestin-1 or -2, KP-triggered GnRH secretion is significantly diminished. Based on these findings, we hypothesized that ablation of Gαq/11 in GnRH neurons would diminish but not completely block KP-triggered GnRH secretion and that Gαq/11-independent GnRH secretion would be sufficient to maintain fertility. To test this, Gnaq (encodes Gαq) was selectively inactivated in the GnRH neurons of global Gna11 (encodes Gα11)-null mice by crossing Gnrh-Cre and Gnaq(fl/fl);Gna11(-/-) mice. Experimental Gnaq(fl/fl);Gna11(-/-);Gnrh-Cre (Gnaq(d/d)) and control Gnaq(fl/fl);Gna11(-/-) (Gnaq(fl/fl)) littermate mice were generated and subjected to reproductive profiling. This process revealed that testicular development and spermatogenesis, preputial separation, and anogenital distance in males and day of vaginal opening and of first estrus in females were significantly less affected in Gnaq(d/d) mice than in previously characterized Kiss1r(-/-) or Kiss1r(d/d) mice. Additionally, Gnaq(d/d) males were subfertile, and although Gnaq(d/d) females did not ovulate spontaneously, they responded efficiently to a single dose of gonadotropins. Finally, KP stimulation triggered a significant increase in gonadotropins and testosterone levels in Gnaq(d/d) mice. We therefore conclude that the milder reproductive phenotypes and maintained responsiveness to KP and gonadotropins reflect Gαq/11-independent GnRH secretion and activation of the neuroendocrine-reproductive axis in Gnaq(d/d) mice. SIGNIFICANCE STATEMENT: The gonadotropin-releasing hormone (GnRH) is the master regulator of fertility. Over the last decade, several studies have established that the KISS1 receptor, KISS1R, is a potent trigger of GnRH secretion and inactivation of KISS1R on the GnRH neuron results in infertility. While KISS1R is best understood as a Gαq/11-coupled receptor, we previously demonstrated that it could couple to and signal via non-Gαq/11-coupled pathways. The present study confirms these findings and, more importantly, while it establishes Gαq/11-coupled signaling as a major conduit of GnRH secretion, it also uncovers a significant role for non-Gαq/11-coupled signaling in potentiating reproductive development and function. This study further suggests that by augmenting signaling via these pathways, GnRH secretion can be enhanced to treat some forms of infertility.


Subject(s)
GTP-Binding Protein alpha Subunits/deficiency , Gonadotropin-Releasing Hormone/physiology , Hypogonadism/physiopathology , Infertility, Female/physiopathology , Infertility, Male/physiopathology , Animals , Blastocyst/pathology , Embryonic Development , Female , GTP-Binding Protein alpha Subunits/physiology , Gene Expression Profiling , Genitalia, Female/pathology , Genitalia, Female/physiopathology , Genitalia, Male/pathology , Genitalia, Male/physiopathology , Gonadal Steroid Hormones/metabolism , Gonadotropin-Releasing Hormone/antagonists & inhibitors , Gonadotropins, Pituitary/metabolism , Gonadotropins, Pituitary/pharmacology , Hypogonadism/genetics , Hypogonadism/pathology , Hypothalamo-Hypophyseal System/physiopathology , Hypothalamus/pathology , Infertility, Female/embryology , Infertility, Female/genetics , Infertility, Male/embryology , Infertility, Male/genetics , Kisspeptins/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/metabolism , Oligopeptides/pharmacology , Ovariectomy , Ovulation/drug effects , Peptide Fragments/pharmacology , Peptides/pharmacology , Phenotype , Receptors, G-Protein-Coupled , Receptors, Kisspeptin-1 , Spermatogenesis
4.
Endocrinology ; 156(2): 627-37, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25422875

ABSTRACT

Tachykinins are comprised of the family of related peptides, substance P (SP), neurokinin A (NKA), and neurokinin B (NKB). NKB has emerged as regulator of kisspeptin release in the arcuate nucleus (ARC), whereas the roles of SP and NKA in reproduction remain unknown. This work explores the roles of SP and NKA in the central regulation of GnRH release. First, central infusion of specific agonists for the receptors of SP (neurokinin receptor 1, NK1R), NKA (NK2R) and NKB (NK3R) each induced gonadotropin release in adult male and ovariectomized, estradiol-replaced female mice, which was absent in Kiss1r(-/-) mice, indicating a kisspeptin-dependent action. The NK2R agonist, however, decreased LH release in ovariectomized-sham replaced females, as documented for NK3R agonists but in contrast to the NK1R agonist, which further increased LH release. Second, Tac1 (encoding SP and NKA) expression in the ARC and ventromedial nucleus was inhibited by circulating estradiol but did not colocalize with Kiss1 mRNA. Third, about half of isolated ARC Kiss1 neurons expressed Tacr1 (NK1R) and 100% Tacr3 (NK3R); for anteroventral-periventricular Kiss1 neurons and GnRH neurons, approximately one-fourth expressed Tacr1 and one-tenth Tacr3; Tacr2 (NK2R) expression was absent in all cases. Overall, these results identify a potent regulation of gonadotropin release by the SP/NK1R and NKA/NK2R systems in the presence of kisspeptin-Kiss1r signaling, indicating that they may, along with NKB/NK3R, control GnRH release, at least in part through actions on Kiss1 neurons.


Subject(s)
Hypothalamus/metabolism , Neurokinin A/metabolism , Reproduction , Substance P/metabolism , Animals , Estradiol/metabolism , Female , Follicle Stimulating Hormone/metabolism , Gonadotropin-Releasing Hormone/metabolism , Kisspeptins/metabolism , Luteinizing Hormone/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Receptors, G-Protein-Coupled/metabolism , Receptors, Kisspeptin-1 , Receptors, Tachykinin/agonists
5.
N Engl J Med ; 368(26): 2467-75, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23738509

ABSTRACT

BACKGROUND: The onset of puberty is first detected as an increase in pulsatile secretion of gonadotropin-releasing hormone (GnRH). Early activation of the hypothalamic-pituitary-gonadal axis results in central precocious puberty. The timing of pubertal development is driven in part by genetic factors, but only a few, rare molecular defects associated with central precocious puberty have been identified. METHODS: We performed whole-exome sequencing in 40 members of 15 families with central precocious puberty. Candidate variants were confirmed with Sanger sequencing. We also performed quantitative real-time polymerase-chain-reaction assays to determine levels of messenger RNA (mRNA) in the hypothalami of mice at different ages. RESULTS: We identified four novel heterozygous mutations in MKRN3, the gene encoding makorin RING-finger protein 3, in 5 of the 15 families; both sexes were affected. The mutations included three frameshift mutations, predicted to encode truncated proteins, and one missense mutation, predicted to disrupt protein function. MKRN3 is a paternally expressed, imprinted gene located in the Prader-Willi syndrome critical region (chromosome 15q11-q13). All affected persons inherited the mutations from their fathers, a finding that indicates perfect segregation with the mode of inheritance expected for an imprinted gene. Levels of Mkrn3 mRNA were high in the arcuate nucleus of prepubertal mice, decreased immediately before puberty, and remained low after puberty. CONCLUSIONS: Deficiency of MKRN3 causes central precocious puberty in humans. (Funded by the National Institutes of Health and others.).


Subject(s)
Frameshift Mutation , Mutation, Missense , Puberty, Precocious/genetics , Ribonucleoproteins/genetics , Animals , Arcuate Nucleus of Hypothalamus/chemistry , Child , Child, Preschool , Exome , Female , Genetic Association Studies , Heterozygote , Humans , Hypothalamus/metabolism , Male , Mice , Pedigree , RNA, Messenger/analysis , Ribonucleoproteins/deficiency , Sequence Analysis, DNA , Ubiquitin-Protein Ligases
SELECTION OF CITATIONS
SEARCH DETAIL