Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Stem Cells ; 40(12): 1107-1121, 2022 12 31.
Article in English | MEDLINE | ID: mdl-36153707

ABSTRACT

Hypothalamic gonadotropin-releasing hormone (GnRH) neurons lay the foundation for human development and reproduction; however, the critical cell populations and the entangled mechanisms underlying the development of human GnRH neurons remain poorly understood. Here, by using our established human pluripotent stem cell-derived GnRH neuron model, we decoded the cellular heterogeneity and differentiation trajectories at the single-cell level. We found that a glutamatergic neuron population, which generated together with GnRH neurons, showed similar transcriptomic properties with olfactory sensory neuron and provided the migratory path for GnRH neurons. Through trajectory analysis, we identified a specific gene module activated along the GnRH neuron differentiation lineage, and we examined one of the transcription factors, DLX5, expression in human fetal GnRH neurons. Furthermore, we found that Wnt inhibition could increase DLX5 expression and improve the GnRH neuron differentiation efficiency through promoting neurogenesis and switching the differentiation fates of neural progenitors into glutamatergic neurons/GnRH neurons. Our research comprehensively reveals the dynamic cell population transition and gene regulatory network during GnRH neuron differentiation.


Subject(s)
Gonadotropin-Releasing Hormone , Pluripotent Stem Cells , Humans , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Wnt Signaling Pathway/genetics , Neurons/metabolism , Cell Differentiation/genetics , Pluripotent Stem Cells/metabolism
2.
Glia ; 66(1): 94-107, 2018 01.
Article in English | MEDLINE | ID: mdl-28887860

ABSTRACT

Radial glial cells play an essential role through their function as guides for neuronal migration during development. Disruption of metabotropic glutamate receptor 5 (mGluR5) function retards the growth of radial glial processes in vitro. Neuregulins (NRG) are activated by proteolytic cleavage and regulate (radial) glial maintenance via ErbB3/ErbB4 receptors. We show here that blocking ErbB4 disrupts radial process extension. Soluble NRG acting on ErbB4 receptors is able to promote radial process extension in particular where process elongation has been impeded by blockade of mGluR5, the nonselective cation channel canonical transient receptor potential 3 (TRPC3), or matrix metalloproteases (MMP). NRG does not restore retarded process growth caused by ErbB4 blockade. Stimulation of muscarinic receptors restores process elongation due to mGluR5 blockade but not that caused by TRPC3, MMP or ErbB4 blockade suggesting that muscarinic receptors can replace mGluR5 with respect to radial process extension. Additionally, NRG/ErbB4 causes Ca2+ mobilization in a population of cells through cooperation with ErbB1 receptors. Our results indicate that mGluR5 promotes radial process growth via NRG activation by a mechanism involving TRPC3 channels and MMPs. Thus neurotransmitters acting on G-protein coupled receptors could play a central role in the maintenance of the radial glial scaffold through activation of NRG/ErbB4 signaling.


Subject(s)
Ependymoglial Cells/drug effects , Glutamic Acid/pharmacology , Neuregulins/metabolism , Receptor, ErbB-4/metabolism , Receptor, Metabotropic Glutamate 5/metabolism , Signal Transduction/drug effects , TRPC Cation Channels/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Embryo, Mammalian , Ependymoglial Cells/metabolism , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Lateral Ventricles/cytology , Mice , Mice, Inbred C57BL , Neuregulins/genetics , RNA, Messenger/metabolism , Receptor, ErbB-4/genetics , Receptor, Metabotropic Glutamate 5/genetics , Signal Transduction/physiology , TRPC Cation Channels/genetics
3.
Eur J Endocrinol ; 189(2): 271-280, 2023 Aug 02.
Article in English | MEDLINE | ID: mdl-37619992

ABSTRACT

OBJECTIVE: Congenital hypogonadotropic hypogonadism (CHH) is a rare, genetically heterogeneous reproductive disorder caused by gonadotropin-releasing hormone (GnRH) deficiency. Approximately half of CHH patients also have decreased or absent sense of smell, that is, Kallmann syndrome (KS). We describe a patient with White-Sutton syndrome (developmental delay and autism spectrum disorder) and KS due to a heterozygous de novo mutation in POGZ (c.2857C>T, p.(Gln953*)), a gene encoding pogo transposable element derived with zinc finger domain, which acts as a transcriptomic regulator of neuronal networks. DESIGN AND METHODS: We modeled the role of POGZ in CHH by generating 2 clonal human pluripotent stem cell lines with CRISPR/Cas9, carrying either the heterozygous patient mutation (H11 line) or a homozygous mutation (c.2803-2906del; p.E935Kfs*7 encoding a truncated POGZ protein; F6del line). RESULTS: During the differentiation to GnRH neurons, neural progenitors derived from F6del line displayed severe proliferation defect, delayed wound-healing capacity, downregulation of intermediate progenitor neuron genes TBR1 and TBR2, and immature neuron markers PAX6 and TUBB3 and gave rise to fewer neurons with shorter neurites and less neurite branch points compared to the WT and H11 lines (P < .005). Both lines, however, could be successfully differentiated to GnRH neurons. CONCLUSIONS: In conclusion, this is the first report on the overlap between White-Sutton syndrome and CHH. POGZ mutations do not hinder GnRH neuron formation but may cause CHH/KS by affecting the size and motility of the anterior neural progenitor pool and neurite outgrowth.


Subject(s)
Autism Spectrum Disorder , Kallmann Syndrome , Humans , Kallmann Syndrome/genetics , Neurons , Gonadotropin-Releasing Hormone , Mutation/genetics
4.
Dis Model Mech ; 15(8)2022 08 01.
Article in English | MEDLINE | ID: mdl-35833364

ABSTRACT

Fibroblast growth factor 8 (FGF8), acting through the fibroblast growth factor receptor 1 (FGFR1), has an important role in the development of gonadotropin-releasing hormone-expressing neurons (GnRH neurons). We hypothesized that FGF8 regulates differentiation of human GnRH neurons in a time- and dose-dependent manner via FGFR1. To investigate this further, human pluripotent stem cells were differentiated during 10 days of dual-SMAD inhibition into neural progenitor cells, followed either by treatment with FGF8 at different concentrations (25 ng/ml, 50 ng/ml or 100 ng/ml) for 10 days or by treatment with 100 ng/ml FGF8 for different durations (2, 4, 6 or 10 days); cells were then matured through DAPT-induced inhibition of Notch signaling for 5 days into GnRH neurons. FGF8 induced expression of GNRH1 in a dose-dependent fashion and the duration of FGF8 exposure correlated positively with gene expression of GNRH1 (P<0.05, Rs=0.49). However, cells treated with 100 ng/ml FGF8 for 2 days induced the expression of genes, such as FOXG1, ETV5 and SPRY2, and continued FGF8 treatment induced the dynamic expression of several other genes. Moreover, during exposure to FGF8, FGFR1 localized to the cell surface and its specific inhibition with the FGFR1 inhibitor PD166866 reduced expression of GNRH1 (P<0.05). In neurons, FGFR1 also localized to the nucleus. Our results suggest that dose- and time-dependent FGF8 signaling via FGFR1 is indispensable for human GnRH neuron ontogeny. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Gonadotropin-Releasing Hormone , Receptor, Fibroblast Growth Factor, Type 1 , Fibroblast Growth Factor 8/metabolism , Fibroblast Growth Factor 8/pharmacology , Forkhead Transcription Factors/metabolism , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/pharmacology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis , Neurons/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism
5.
Eur J Endocrinol ; 185(1): 57-66, 2021 May 21.
Article in English | MEDLINE | ID: mdl-33909591

ABSTRACT

Patients with deletions on chromosome 9q31.2 may exhibit delayed puberty, craniofacial phenotype including cleft lip/palate, and olfactory bulb hypoplasia. We report a patient with congenital HH with anosmia (Kallmann syndrome, KS) and a de novo 2.38 Mb heterozygous deletion in 9q31.2. The deletion breakpoints (determined with whole-genome linked-read sequencing) were in the FKTN gene (9:108,331,353) and in a non-coding area (9:110,707,332) (hg19). The deletion encompassed six protein-coding genes (FKTN, ZNF462, TAL2, TMEM38B, RAD23B, and KLF4). ZNF462 haploinsufficiency was consistent with the patient's Weiss-Kruszka syndrome (craniofacial phenotype, developmental delay, and sensorineural hearing loss), but did not explain his KS. In further analyses, he did not carry rare sequence variants in 32 known KS genes in whole-exome sequencing and displayed no aberrant splicing of 15 KS genes that were expressed in peripheral blood leukocyte transcriptome. The deletion was 1.8 Mb upstream of a KS candidate gene locus (PALM2AKAP2) but did not suppress its expression. In conclusion, this is the first report of a patient with Weiss-Kruszka syndrome and KS. We suggest that patients carrying a microdeletion in 9q31.2 should be evaluated for the presence of KS and KS-related features.


Subject(s)
Craniofacial Abnormalities/genetics , DNA-Binding Proteins/genetics , Developmental Disabilities/genetics , Hearing Loss, Sensorineural/genetics , Heart Septal Defects/genetics , Kallmann Syndrome/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Transcription Factors/genetics , Adolescent , Basic Helix-Loop-Helix Transcription Factors/genetics , Chromosomes, Human, Pair 9/genetics , Craniofacial Abnormalities/complications , DNA Repair Enzymes/genetics , Developmental Disabilities/complications , Gene Deletion , Haploinsufficiency , Hearing Loss, Sensorineural/complications , Heart Septal Defects/complications , Humans , Ion Channels/genetics , Kallmann Syndrome/complications , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Male , Membrane Proteins/metabolism , Neoplasm Proteins/genetics , Sequence Analysis, RNA , Syndrome , Exome Sequencing
6.
Dis Model Mech ; 13(3)2020 03 13.
Article in English | MEDLINE | ID: mdl-31996360

ABSTRACT

Gonadotropin-releasing hormone (GnRH) neurons provide a fundamental signal for the onset of puberty and subsequent reproductive functions by secretion of gonadotropin-releasing hormone. Their disrupted development or function leads to congenital hypogonadotropic hypogonadism (CHH). To model the development of human GnRH neurons, we generated a stable GNRH1-TdTomato reporter cell line in human pluripotent stem cells (hPSCs) using CRISPR-Cas9 genome editing. RNA-sequencing of the reporter clone, differentiated into GnRH neurons by dual SMAD inhibition and FGF8 treatment, revealed 6461 differentially expressed genes between progenitors and GnRH neurons. Expression of the transcription factor ISL1, one of the top 50 most upregulated genes in the TdTomato-expressing GnRH neurons, was confirmed in 10.5 gestational week-old human fetal GnRH neurons. Among the differentially expressed genes, we detected 15 genes that are implicated in CHH and several genes that are implicated in human puberty timing. Finally, FGF8 treatment in the neuronal progenitor pool led to upregulation of 37 genes expressed both in progenitors and in TdTomato-expressing GnRH neurons, which suggests upstream regulation of these genes by FGF8 signaling during GnRH neuron differentiation. These results illustrate how hPSC-derived human GnRH neuron transcriptomic analysis can be utilized to dissect signaling pathways and gene regulatory networks involved in human GnRH neuron development.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Genes, Reporter , Gonadotropin-Releasing Hormone/metabolism , Neurons/metabolism , Pluripotent Stem Cells/metabolism , Transcriptome/genetics , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Cell Line , Fetus/cytology , Fibroblast Growth Factor 8/pharmacology , Humans , Hypogonadism/genetics , LIM-Homeodomain Proteins/metabolism , Neurons/drug effects , Pluripotent Stem Cells/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
7.
Article in English | MEDLINE | ID: mdl-30800097

ABSTRACT

Paternally-inherited loss-of-function mutations in makorin ring finger protein 3 gene (MKRN3) underlie central precocious puberty. To investigate the puberty-related mechanism(s) of MKRN3 in humans, we generated two distinct bi-allelic MKRN3 knock-out human pluripotent stem cell lines, Del 1 and Del 2, and differentiated them into GNRH1-expressing neurons. Both Del 1 and Del 2 clones could be differentiated into neuronal progenitors and GNRH1-expressing neurons, however, the relative expression of GNRH1 did not differ from wild type cells (P = NS). Subsequently, we investigated stable and dynamic protein-protein interaction (PPI) partners of MKRN3 by stably expressing it in HEK cells followed by mass spectrometry analyses. We found 81 high-confidence novel protein interaction partners, which are implicated in cellular processes such as insulin signaling, RNA metabolism and cell-cell adhesion. Of the identified interactors, 20 have been previously implicated in puberty timing. In conclusion, our stem cell model for generation of GNRH1-expressing neurons did not offer mechanistic insight for the role of MKRN3 in puberty initiation. The PPI data, however, indicate that MKRN3 may regulate puberty by interacting with other puberty-related proteins. Further studies are required to elucidate the possible mechanisms and outcomes of these interactions.

8.
Stem Cell Reports ; 7(2): 149-57, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27426041

ABSTRACT

Gonadotropin-releasing hormone (GnRH) neurons regulate human puberty and reproduction. Modeling their development and function in vitro would be of interest for both basic research and clinical translation. Here, we report a three-step protocol to differentiate human pluripotent stem cells (hPSCs) into GnRH-secreting neurons. Firstly, hPSCs were differentiated to FOXG1, EMX2, and PAX6 expressing anterior neural progenitor cells (NPCs) by dual SMAD inhibition. Secondly, NPCs were treated for 10 days with FGF8, which is a key ligand implicated in GnRH neuron ontogeny, and finally, the cells were matured with Notch inhibitor to bipolar TUJ1-positive neurons that robustly expressed GNRH1 and secreted GnRH decapeptide into the culture medium. The protocol was reproducible both in human embryonic stem cells and induced pluripotent stem cells, and thus provides a translational tool for investigating the mechanisms of human puberty and its disorders.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Neurons/cytology , Neurons/metabolism , Pluripotent Stem Cells/cytology , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Fibroblast Growth Factor 8/pharmacology , Forkhead Transcription Factors/metabolism , Humans , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurons/drug effects , Nose/cytology , Pluripotent Stem Cells/metabolism , Receptors, Notch/metabolism , Smad Proteins/antagonists & inhibitors , Smad Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL