Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Genome Med ; 16(1): 75, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38822427

ABSTRACT

BACKGROUND: Congenital hypopituitarism (CH) and its associated syndromes, septo-optic dysplasia (SOD) and holoprosencephaly (HPE), are midline defects that cause significant morbidity for affected people. Variants in 67 genes are associated with CH, but a vast majority of CH cases lack a genetic diagnosis. Whole exome and whole genome sequencing of CH patients identifies sequence variants in genes known to cause CH, and in new candidate genes, but many of these are variants of uncertain significance (VUS). METHODS: The International Mouse Phenotyping Consortium (IMPC) is an effort to establish gene function by knocking-out all genes in the mouse genome and generating corresponding phenotype data. We used mouse embryonic imaging data generated by the Deciphering Mechanisms of Developmental Disorders (DMDD) project to screen 209 embryonic lethal and sub-viable knockout mouse lines for pituitary malformations. RESULTS: Of the 209 knockout mouse lines, we identified 51 that have embryonic pituitary malformations. These genes not only represent new candidates for CH, but also reveal new molecular pathways not previously associated with pituitary organogenesis. We used this list of candidate genes to mine whole exome sequencing data of a cohort of patients with CH, and we identified variants in two unrelated cases for two genes, MORC2 and SETD5, with CH and other syndromic features. CONCLUSIONS: The screening and analysis of IMPC phenotyping data provide proof-of-principle that recessive lethal mouse mutants generated by the knockout mouse project are an excellent source of candidate genes for congenital hypopituitarism in children.


Subject(s)
Hypopituitarism , Mice, Knockout , Pituitary Gland , Hypopituitarism/genetics , Animals , Humans , Pituitary Gland/metabolism , Pituitary Gland/abnormalities , Pituitary Gland/pathology , Mice , Phenotype , Female , Male , Disease Models, Animal , Exome Sequencing , Septo-Optic Dysplasia/genetics
2.
bioRxiv ; 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38746430

ABSTRACT

Myelin regulatory factor (Myrf) is a critical transcription factor in early retinal and retinal pigment epithelial development, and human variants in MYRF are a cause for nanophthalmos. Single cell RNA sequencing (scRNAseq) was performed on Myrf conditional knockout mice ( Rx>Cre Myrf fl/fl ) at 3 developmental timepoints. Myrf was expressed specifically in the RPE, and expression was abrogated in Rx>Cre Myrf fl/fl eyes. scRNAseq analysis revealed a loss of RPE cells at all timepoints resulting from cell death. GO-term analysis in the RPE revealed downregulation of melanogenesis and anatomic structure morphogenesis pathways, which were supported by electron microscopy and histologic analysis. Novel structural target genes including Ermn and Upk3b , along with macular degeneration and inherited retinal disease genes were identified as downregulated, and a strong upregulation of TGFß/BMP signaling and effectors was observed. Regulon analysis placed Myrf downstream of Pax6 and Mitf and upstream of Sox10 in RPE differentiation. Together, these results suggest a strong role for Myrf in the RPE maturation by regulating melanogenesis, cell survival, and cell structure, in part acting through suppression of TGFß signaling and activation of Sox10 . SUMMARY STATEMENT: Myrf regulates RPE development, melanogenesis, and is important for cell structure and survival, in part through regulation of Ermn , Upk3b and Sox10, and BMP/TGFb signaling.

3.
Nat Rev Endocrinol ; 20(2): 77-92, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38102391

ABSTRACT

Pituitary cells that express the transcription factor SOX2 are stem cells because they can self-renew and differentiate into multiple pituitary hormone-producing cell types as organoids. Wounding and physiological challenges can activate pituitary stem cells, but cell numbers are not fully restored, and the ability to mobilize stem cells decreases with increasing age. The basis of these limitations is still unknown. The regulation of stem cell quiescence and activation involves many different signalling pathways, including those mediated by WNT, Hippo and several cytokines; more research is needed to understand the interactions between these pathways. Pituitary organoids can be formed from human or mouse embryonic stem cells, or from human induced pluripotent stem cells. Human pituitary organoid transplantation is sufficient to induce corticosterone release in hypophysectomized mice, raising the possibility of therapeutic applications. Today, pituitary organoids have the potential to assess the role of individual genes and genetic variants on hormone production ex vivo, providing an important tool for the advancement of exciting frontiers in pituitary stem cell biology and pituitary organogenesis. In this article, we provide an overview of notable discoveries in pituitary stem cell function and highlight important areas for future research.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Animals , Mice , Induced Pluripotent Stem Cells/metabolism , Pituitary Gland/metabolism , Transcription Factors/metabolism , Signal Transduction , Cell Differentiation
4.
Endocrinology ; 164(6)2023 04 17.
Article in English | MEDLINE | ID: mdl-37183548

ABSTRACT

The pituitary gland regulates growth, metabolism, reproduction, the stress response, uterine contractions, lactation, and water retention. It secretes hormones in response to hypothalamic input, end organ feedback, and diurnal cues. The mechanisms by which pituitary stem cells are recruited to proliferate, maintain quiescence, or differentiate into specific cell types, especially thyrotropes, are not well understood. We used single-cell RNA sequencing in juvenile P7 mouse pituitary cells to identify novel factors in pituitary cell populations, with a focus on thyrotropes and rare subtypes. We first observed cells coexpressing markers of both thyrotropes and gonadotropes, such as Pou1f1 and Nr5a1. This was validated in vivo by both immunohistochemistry and lineage tracing of thyrotropes derived from Nr5a1-Cre; mTmG mice and demonstrates that Nr5a1-progenitors give rise to a proportion of thyrotropes during development. Our data set also identifies novel factors expressed in pars distalis and pars tuberalis thyrotropes, including the Shox2b isoform in all thyrotropes and Sox14 specifically in Pou1f1-negative pars tuberalis thyrotropes. We have therefore used single-cell transcriptomics to determine a novel developmental trajectory for thyrotropes and potential novel regulators of thyrotrope populations.


Subject(s)
Pituitary Diseases , Pituitary Gland, Anterior , Pregnancy , Female , Mice , Animals , Thyrotropin/metabolism , Pituitary Gland/metabolism , Transcription Factors/metabolism , Pituitary Diseases/metabolism , Immunohistochemistry , Pituitary Gland, Anterior/metabolism , SOXB2 Transcription Factors/metabolism
5.
Hum Mol Genet ; 32(3): 367-385, 2023 01 13.
Article in English | MEDLINE | ID: mdl-35951005

ABSTRACT

Congenital hypopituitarism is a genetically heterogeneous condition that is part of a spectrum disorder that can include holoprosencephaly. Heterozygous mutations in SIX3 cause variable holoprosencephaly in humans and mice. We identified two children with neonatal hypopituitarism and thin pituitary stalk who were doubly heterozygous for rare, likely deleterious variants in the transcription factors SIX3 and POU1F1. We used genetically engineered mice to understand the disease pathophysiology. Pou1f1 loss-of-function heterozygotes are unaffected; Six3 heterozygotes have pituitary gland dysmorphology and incompletely ossified palate; and the Six3+/-; Pou1f1+/dw double heterozygote mice have a pronounced phenotype, including pituitary growth through the palate. The interaction of Pou1f1 and Six3 in mice supports the possibility of digenic pituitary disease in children. Disruption of Six3 expression in the oral ectoderm completely ablated anterior pituitary development, and deletion of Six3 in the neural ectoderm blocked the development of the pituitary stalk and both anterior and posterior pituitary lobes. Six3 is required in both oral and neural ectodermal tissues for the activation of signaling pathways and transcription factors necessary for pituitary cell fate. These studies clarify the mechanism of SIX3 action in pituitary development and provide support for a digenic basis for hypopituitarism.


Subject(s)
Holoprosencephaly , Hypopituitarism , Child , Humans , Heterozygote , Hypopituitarism/genetics , Transcription Factors/genetics , Mutation , Pituitary Hormones/genetics , Transcription Factor Pit-1/genetics
6.
J Clin Invest ; 130(8): 4501-4515, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32453714

ABSTRACT

The transcription factor ISL1 is expressed in pituitary gland stem cells and the thyrotrope and gonadotrope lineages. Pituitary-specific Isl1 deletion causes hypopituitarism with increased stem cell apoptosis, reduced differentiation of thyrotropes and gonadotropes, and reduced body size. Conditional Isl1 deletion causes development of multiple Rathke's cleft-like cysts, with 100% penetrance. Foxa1 and Foxj1 are abnormally expressed in the pituitary gland and associated with a ciliogenic gene-expression program in the cysts. We confirmed expression of FOXA1, FOXJ1, and stem cell markers in human Rathke's cleft cyst tissue, but not craniopharyngiomas, which suggests these transcription factors are useful, pathological markers for diagnosis of Rathke's cleft cysts. These studies support a model whereby expression of ISL1 in pituitary progenitors drives differentiation into thyrotropes and gonadotropes and without it, activation of FOXA1 and FOXJ1 permits development of an oral epithelial cell fate with mucinous cysts. This pituitary-specific Isl1 mouse knockout sheds light on the etiology of Rathke's cleft cysts and the role of ISL1 in normal pituitary development.


Subject(s)
Central Nervous System Cysts/metabolism , Gene Deletion , LIM-Homeodomain Proteins/deficiency , Neoplasm Proteins/deficiency , Pituitary Gland/metabolism , Stem Cells/metabolism , Transcription Factors/deficiency , Animals , Central Nervous System Cysts/genetics , Central Nervous System Cysts/pathology , LIM-Homeodomain Proteins/metabolism , Mice , Mice, Knockout , Neoplasm Proteins/metabolism , Pituitary Gland/pathology , Stem Cells/pathology , Transcription Factors/metabolism
7.
PLoS Genet ; 15(5): e1008130, 2019 05.
Article in English | MEDLINE | ID: mdl-31048900

ABSTRACT

Nanophthalmos is a rare, potentially devastating eye condition characterized by small eyes with relatively normal anatomy, a high hyperopic refractive error, and frequent association with angle closure glaucoma and vision loss. The condition constitutes the extreme of hyperopia or farsightedness, a common refractive error that is associated with strabismus and amblyopia in children. NNO1 was the first mapped nanophthalmos locus. We used combined pooled exome sequencing and strong linkage data in the large family used to map this locus to identify a canonical splice site alteration upstream of the last exon of the gene encoding myelin regulatory factor (MYRF c.3376-1G>A), a membrane bound transcription factor that undergoes autoproteolytic cleavage for nuclear localization. This variant produced a stable RNA transcript, leading to a frameshift mutation p.Gly1126Valfs*31 in the C-terminus of the protein. In addition, we identified an early truncating MYRF frameshift mutation, c.769dupC (p.S264QfsX74), in a patient with extreme axial hyperopia and syndromic features. Myrf conditional knockout mice (CKO) developed depigmentation of the retinal pigment epithelium (RPE) and retinal degeneration supporting a role of this gene in retinal and RPE development. Furthermore, we demonstrated the reduced expression of Tmem98, another known nanophthalmos gene, in Myrf CKO mice, and the physical interaction of MYRF with TMEM98. Our study establishes MYRF as a nanophthalmos gene and uncovers a new pathway for eye growth and development.


Subject(s)
Glaucoma, Angle-Closure/genetics , Hyperopia/genetics , Membrane Proteins/genetics , Microphthalmos/genetics , Retinal Degeneration/genetics , Transcription Factors/genetics , Adult , Animals , Child , Child, Preschool , Exons , Family , Female , Frameshift Mutation/genetics , Genetic Variation/genetics , Glaucoma, Angle-Closure/metabolism , Humans , Hyperopia/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microphthalmos/metabolism , Middle Aged , Pedigree , RNA Splice Sites/genetics , Refractive Errors/genetics , Transcription Factors/metabolism
8.
Endocrinology ; 159(12): 3910-3924, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30335147

ABSTRACT

Transcription factors and signaling pathways that regulate stem cells and specialized hormone-producing cells in the pituitary gland have been the subject of intense study and have yielded a mechanistic understanding of pituitary organogenesis and disease. However, the regulation of stem cell proliferation and differentiation, the heterogeneity among specialized hormone-producing cells, and the role of nonendocrine cells in the gland remain important, unanswered questions. Recent advances in single-cell RNA sequencing (scRNAseq) technologies provide new avenues to address these questions. We performed scRNAseq on ∼13,663 cells pooled from six whole pituitary glands of 7-week-old C57BL/6 male mice. We identified pituitary endocrine and stem cells in silico, as well as other support cell types such as endothelia, connective tissue, and red and white blood cells. Differential gene expression analyses identify known and novel markers of pituitary endocrine and stem cell populations. We demonstrate the value of scRNAseq by in vivo validation of a novel gonadotrope-enriched marker, Foxp2. We present novel scRNAseq data of in vivo pituitary tissue, including data from agnostic clustering algorithms that suggest the presence of a somatotrope subpopulation enriched in sterol/cholesterol synthesis genes. Additionally, we show that incomplete transcriptome annotation can cause false negatives on some scRNAseq platforms that only generate 3' transcript end sequences, and we use in vivo data to recover reads of the pituitary transcription factor Prop1. Ultimately, scRNAseq technologies represent a significant opportunity to address long-standing questions regarding the development and function of the different populations of the pituitary gland throughout life.


Subject(s)
Biomarkers/analysis , Pituitary Gland/cytology , Pituitary Gland/metabolism , Pituitary Hormones/metabolism , Single-Cell Analysis/methods , Stem Cells/classification , Stem Cells/metabolism , Animals , Biomarkers/metabolism , Cell Differentiation/genetics , Cell Separation/methods , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Organogenesis/genetics , Pituitary Gland/embryology , Pituitary Hormones/genetics , Sequence Analysis, RNA/methods , Stem Cells/cytology , Transcriptome
9.
Mol Cell Endocrinol ; 445: 14-26, 2017 04 15.
Article in English | MEDLINE | ID: mdl-27650955

ABSTRACT

The anterior pituitary gland is comprised of specialized cell-types that produce and secrete polypeptide hormones in response to hypothalamic input and feedback from target organs. These specialized cells arise from stem cells that express SOX2 and the pituitary transcription factor PROP1, which is necessary to establish the stem cell pool and promote an epithelial to mesenchymal-like transition, releasing progenitors from the niche. The adult anterior pituitary responds to physiological challenge by mobilizing the SOX2-expressing progenitor pool and producing additional hormone-producing cells. Knowledge of the role of signaling pathways and extracellular matrix components in these processes may lead to improvements in the efficiency of differentiation of embryonic stem cells or induced pluripotent stem cells into hormone producing cells in vitro. Advances in our basic understanding of pituitary stem cell regulation and differentiation may lead to improved diagnosis and treatment for patients with hypopituitarism.


Subject(s)
Homeodomain Proteins/metabolism , Pituitary Gland, Anterior/cytology , SOXB1 Transcription Factors/metabolism , Stem Cells/cytology , Animals , Cell Differentiation , Epithelial-Mesenchymal Transition , Extracellular Matrix/metabolism , Humans , Pituitary Gland, Anterior/metabolism , Signal Transduction , Stem Cells/metabolism
10.
Endocr Rev ; 37(6): 636-675, 2016 12.
Article in English | MEDLINE | ID: mdl-27828722

ABSTRACT

The genetic basis for combined pituitary hormone deficiency (CPHD) is complex, involving 30 genes in a variety of syndromic and nonsyndromic presentations. Molecular diagnosis of this disorder is valuable for predicting disease progression, avoiding unnecessary surgery, and family planning. We expect that the application of high throughput sequencing will uncover additional contributing genes and eventually become a valuable tool for molecular diagnosis. For example, in the last 3 years, six new genes have been implicated in CPHD using whole-exome sequencing. In this review, we present a historical perspective on gene discovery for CPHD and predict approaches that may facilitate future gene identification projects conducted by clinicians and basic scientists. Guidelines for systematic reporting of genetic variants and assigning causality are emerging. We apply these guidelines retrospectively to reports of the genetic basis of CPHD and summarize modes of inheritance and penetrance for each of the known genes. In recent years, there have been great improvements in databases of genetic information for diverse populations. Some issues remain that make molecular diagnosis challenging in some cases. These include the inherent genetic complexity of this disorder, technical challenges like uneven coverage, differing results from variant calling and interpretation pipelines, the number of tolerated genetic alterations, and imperfect methods for predicting pathogenicity. We discuss approaches for future research in the genetics of CPHD.


Subject(s)
Genomics/methods , Hypopituitarism/genetics , Animals , Humans
11.
Elife ; 52016 06 28.
Article in English | MEDLINE | ID: mdl-27351100

ABSTRACT

Mutations in PROP1 are the most common cause of hypopituitarism in humans; therefore, unraveling its mechanism of action is highly relevant from a therapeutic perspective. Our current understanding of the role of PROP1 in the pituitary gland is limited to the repression and activation of the pituitary transcription factor genes Hesx1 and Pou1f1, respectively. To elucidate the comprehensive PROP1-dependent gene regulatory network, we conducted genome-wide analysis of PROP1 DNA binding and effects on gene expression in mutant mice, mouse isolated stem cells and engineered mouse cell lines. We determined that PROP1 is essential for stimulating stem cells to undergo an epithelial to mesenchymal transition-like process necessary for cell migration and differentiation. Genomic profiling reveals that PROP1 binds to genes expressed in epithelial cells like Claudin 23, and to EMT inducer genes like Zeb2, Notch2 and Gli2. Zeb2 activation appears to be a key step in the EMT process. Our findings identify PROP1 as a central transcriptional component of pituitary stem cell differentiation.


Subject(s)
Cell Differentiation , Epithelial-Mesenchymal Transition , Homeodomain Proteins/metabolism , Stem Cells/physiology , Animals , Cell Line , Mice , Mice, Knockout
12.
PLoS Genet ; 11(10): e1005569, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26496357

ABSTRACT

Skeletal dysplasias are a common, genetically heterogeneous cause of short stature that can result from disruptions in many cellular processes. We report the identification of the lesion responsible for skeletal dysplasia and male infertility in the spontaneous, recessive mouse mutant chagun. We determined that Poc1a, encoding protein of the centriole 1a, is disrupted by the insertion of a processed Cenpw cDNA, which is flanked by target site duplications, suggestive of a LINE-1 retrotransposon-mediated event. Mutant fibroblasts have impaired cilia formation and multipolar spindles. Male infertility is caused by defective spermatogenesis early in meiosis and progressive germ cell loss. Spermatogonial stem cell transplantation studies revealed that Poc1a is essential for normal function of both Sertoli cells and germ cells. The proliferative zone of the growth plate is small and disorganized because chondrocytes fail to re-align after cell division and undergo increased apoptosis. Poc1a and several other genes associated with centrosome function can affect the skeleton and lead to skeletal dysplasias and primordial dwarfisms. This mouse mutant reveals how centrosome dysfunction contributes to defects in skeletal growth and male infertility.


Subject(s)
Cytoskeletal Proteins/genetics , Dwarfism/genetics , Infertility, Male/genetics , Long Interspersed Nucleotide Elements/genetics , Spermatogenesis/genetics , Animals , Cell Cycle Proteins , Centrioles/genetics , Centrosome/metabolism , Chromosomal Proteins, Non-Histone/genetics , Dwarfism/pathology , Humans , Infertility, Male/pathology , Male , Meiosis/genetics , Mice , Proteins/genetics , Proteins/metabolism , Sertoli Cells/metabolism , Spermatogonia/metabolism
13.
Biol Reprod ; 93(5): 121, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26333994

ABSTRACT

Chromatin remodeling influences gene expression in developing and adult organisms. Active and repressive marks of histone methylation dictate the embryonic expression boundaries of developmentally regulated genes, including the Hox gene cluster. Drosophila ash1 (absent, small or homeotic discs 1) gene encodes a histone methyltransferase essential for regulation of Hox gene expression that interacts genetically with other members of the trithorax group (TrxG). While mammalian members of the mixed lineage leukemia (Mll) family of TrxG genes have roles in regulation of Hox gene expression, little is known about the expression and function of the mammalian ortholog of the Drosophila ash1 gene, Ash1-like (Ash1l). Here we report the expression of mouse Ash1l gene in specific structures within various organs and provide evidence that reduced Ash1l expression has tissue-specific effects on mammalian development and adult homeostasis. Mutants exhibit partially penetrant postnatal lethality and failure to thrive. Surviving mutants have growth insufficiency, skeletal transformations, and infertility associated with developmental defects in both male and female reproductive organs. Specifically, expression of Hoxa11 and Hoxd10 are altered in the epididymis of Ash1l mutant males and Hoxa10 is reduced in the uterus of Ash1l mutant females. In summary, we show that the histone methyltransferase Ash1l is important for the development and function of several tissues and for proper expression of homeotic genes in mammals.


Subject(s)
DNA-Binding Proteins/deficiency , Epididymis/abnormalities , Fertility , Transcription Factors/deficiency , Uterus/abnormalities , Alleles , Animals , DNA-Binding Proteins/genetics , Epididymis/metabolism , Female , Genes, Homeobox , Histone-Lysine N-Methyltransferase , Homeodomain Proteins/metabolism , Male , Mice, Inbred C57BL , Transcription Factors/genetics
14.
Mol Endocrinol ; 29(4): 597-612, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25668206

ABSTRACT

Defects in the Lhx4, Lhx3, and Pitx2 genes can cause combined pituitary hormone deficiency and pituitary hypoplasia in both humans and mice. Not much is known about the mechanism underlying hypoplasia in these mutants beyond generally increased cell death and poorly maintained proliferation. We identified both common and unique abnormalities in developmental regulation of key cell cycle regulator gene expression in each of these three mutants. All three mutants exhibit reduced expression of the proliferative marker Ki67 and the transitional marker p57. We discovered that expression of the cyclin-dependent kinase inhibitor 1a (Cdkn1a or p21) is expanded dorsally in the pituitary primordium of both Lhx3 and Lhx4 mutants. Uniquely, Lhx4 mutants exhibit reduced cyclin D1 expression and have auxiliary pouch-like structures. We show evidence for indirect and direct effects of LHX4 on p21 expression in αT3-1 pituitary cells. In summary, Lhx4 is necessary for efficient pituitary progenitor cell proliferation and restriction of p21 expression.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Hypopituitarism/metabolism , LIM-Homeodomain Proteins/metabolism , Pituitary Gland/metabolism , Transcription Factors/metabolism , Animals , Cell Proliferation , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Hypopituitarism/genetics , Hypopituitarism/pathology , LIM-Homeodomain Proteins/genetics , Mice , Mice, Transgenic , Pituitary Gland/pathology , Transcription Factors/genetics , Homeobox Protein PITX2
15.
J Biol Chem ; 290(5): 2699-714, 2015 Jan 30.
Article in English | MEDLINE | ID: mdl-25525274

ABSTRACT

Pituitary gonadotropins follicle-stimulating hormone and luteinizing hormone are heterodimeric glycoproteins expressed in gonadotropes. They act on gonads and promote their development and functions including steroidogenesis and gametogenesis. Although transcriptional regulation of gonadotropin subunits has been well studied, the post-transcriptional regulation of gonadotropin subunits is not well understood. To test if microRNAs regulate the hormone-specific gonadotropin ß subunits in vivo, we deleted Dicer in gonadotropes by a Cre-lox genetic approach. We found that many of the DICER-dependent microRNAs, predicted in silico to bind gonadotropin ß subunit mRNAs, were suppressed in purified gonadotropes of mutant mice. Loss of DICER-dependent microRNAs in gonadotropes resulted in profound suppression of gonadotropin-ß subunit proteins and, consequently, the heterodimeric hormone secretion. In addition to suppression of basal levels, interestingly, the post-gonadectomy-induced rise in pituitary gonadotropin synthesis and secretion were both abolished in mutants, indicating a defective gonadal negative feedback control. Furthermore, mutants lacking Dicer in gonadotropes displayed severely reduced fertility and were rescued with exogenous hormones confirming that the fertility defects were secondary to suppressed gonadotropins. Our studies reveal that DICER-dependent microRNAs are essential for gonadotropin homeostasis and fertility in mice. Our studies also implicate microRNAs in gonadal feedback control of gonadotropin synthesis and secretion. Thus, DICER-dependent microRNAs confer a new layer of transcriptional and post-transcriptional regulation in gonadotropes to orchestrate the hypothalamus-pituitary-gonadal axis physiology.


Subject(s)
DEAD-box RNA Helicases/metabolism , Gonadotrophs/metabolism , Gonadotropins/metabolism , Ribonuclease III/metabolism , Animals , DEAD-box RNA Helicases/genetics , Female , Fertility/genetics , Fertility/physiology , Gonadotropins/genetics , Male , Mice , Mice, Knockout , MicroRNAs/genetics , Rats , Real-Time Polymerase Chain Reaction , Ribonuclease III/genetics
16.
Curr Top Dev Biol ; 106: 1-47, 2013.
Article in English | MEDLINE | ID: mdl-24290346

ABSTRACT

Many aspects of pituitary development have become better understood in the past two decades. The signaling pathways regulating pituitary growth and shape have emerged, and the balancing interactions between the pathways are now appreciated. Markers for multipotent progenitor cells are being identified, and signature transcription factors have been discovered for most hormone-producing cell types. We now realize that pulsatile hormone secretion involves a 3D integration of cellular networks. About a dozen genes are known to cause pituitary hypoplasia when mutated due to their essential roles in pituitary development. Similarly, a few genes are known that predispose to familial endocrine neoplasia, and several genes mutated in sporadic pituitary adenomas are documented. In the next decade, we anticipate gleaning a deeper appreciation of these processes at the molecular level, insight into the development of the hypophyseal portal blood system, and evolution of better therapeutics for congenital and acquired hormone deficiencies and for common craniopharyngiomas and pituitary adenomas.


Subject(s)
Gene Expression Regulation, Developmental , Mutation , Pituitary Diseases/genetics , Pituitary Gland/metabolism , Animals , Humans , Models, Genetic , Pituitary Diseases/metabolism , Pituitary Diseases/physiopathology , Pituitary Gland/growth & development , Pituitary Hormones/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Stem Cells/cytology , Stem Cells/metabolism
17.
Hum Mol Genet ; 22(2): 345-57, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23065701

ABSTRACT

We discovered a new spontaneous mutant allele of Npr2 named peewee (pwe) that exhibits severe disproportionate dwarfism and female infertility. The pwe phenotype is caused by a four base-pair deletion in exon 3 that generates a premature stop codon at codon 313 (L313X). The Npr2(pwe/pwe) mouse is a model for the human skeletal dysplasia acromesomelic dysplasia, Maroteaux type (AMDM). We conducted a thorough analysis of the female reproductive tract and report that the primary cause of Npr2(pwe/pwe) female infertility is premature oocyte meiotic resumption, while the pituitary and uterus appear to be normal. Npr2 is expressed in chondrocytes and osteoblasts. We determined that the loss of Npr2 causes a reduction in the hypertrophic and proliferative zones of the growth plate, but mineralization of skeletal elements is normal. Mutant tibiae have increased levels of the activated form of ERK1/2, consistent with the idea that natriuretic peptide receptor type 2 (NPR2) signaling inhibits the activation of the MEK/ERK mitogen activated protein kinase pathway. Treatment of fetal tibiae explants with mitogen activated protein kinase 1 and 2 inhibitors U0126 and PD325901 rescues the Npr2(pwe/pwe) growth defect, providing a promising foundation for skeletal dysplasia therapeutics.


Subject(s)
Bone Diseases, Developmental/genetics , Mutation , Receptors, Atrial Natriuretic Factor/genetics , Reproduction/genetics , Animals , Base Sequence , Bone Density/genetics , Bone Diseases, Developmental/drug therapy , Bone and Bones/metabolism , Dwarfism/genetics , Female , Genotype , Humans , Infertility, Female/genetics , MAP Kinase Signaling System/drug effects , Male , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phenotype , Phosphorylation/drug effects , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology
18.
Mol Endocrinol ; 24(4): 754-65, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20181723

ABSTRACT

Pituitary hormone deficiency causes short stature in one in 4000 children born and can be caused by mutations in transcription factor genes, including HESX1, PROP1, and POU1F1. HESX1 interacts with a member of the groucho-related gene family, TLE1, through an engrailed homology domain and represses PROP1 activity. Mice with Prop1 deficiency exhibit failed differentiation of the POU1F1 lineage, resulting in lack of TSH, GH, and prolactin. In addition, these mutants exhibit profound pituitary dysmorphology and excess Hesx1 and Tle3 expression. The ability of HESX1 to interact with TLE3 has not been explored previously. We tested the ability of TLE3 to enhance HESX1-mediated repression of PROP1 in cell culture. Both TLE3 and TLE1 repress PROP1 in conjunction with HESX1 with similar efficiencies. TLE1 and TLE3 can each repress PROP1 in the absence of HESX1 via a protein-protein interaction. We tested the functional consequences of ectopic TLE3 and HESX1 expression in transgenic mice by driving constitutive expression in pituitary thyrotrophs and gonadotrophs. Terminal differentiation of these cells was suppressed by HESX1 alone and by TLE3 and HESX1 together but not by TLE3 alone. In summary, we present evidence that HESX1 is a strong repressor that can be augmented by the corepressors TLE1 and TLE3. Our in vitro studies suggest that TLE1 and TLE3 might also play roles independent of HESX1 by interacting with other transcription factors like PROP1.


Subject(s)
Homeodomain Proteins/metabolism , Repressor Proteins/metabolism , Animals , Cell Line , Co-Repressor Proteins , Electrophoretic Mobility Shift Assay , Female , Genotype , Homeodomain Proteins/genetics , Humans , Immunohistochemistry , Immunoprecipitation , In Situ Hybridization , Male , Mice , Mice, Transgenic , Open Reading Frames/genetics , Pregnancy , Proteins/genetics , Proteins/metabolism , Repressor Proteins/genetics
19.
Horm Res ; 71 Suppl 2: 101-15, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19407506

ABSTRACT

Genetic cases of congenital pituitary hormone deficiency are common and many are caused by transcription factor defects. Mouse models with orthologous mutations are invaluable for uncovering the molecular mechanisms that lead to problems in organ development and typical patient characteristics. We are using mutant mice defective in the transcription factors PROP1 and POU1F1 for gene expression profiling to identify target genes for these critical transcription factors and candidates for cases of pituitary hormone deficiency of unknown aetiology. These studies reveal critical roles for Wnt signalling pathways, including the TCF/LEF transcription factors and interacting proteins of the groucho family, bone morphogenetic protein antagonists and targets of notch signalling. Current studies are investigating the roles of novel homeobox genes and pathways that regulate the transition from proliferation to differentiation, cell adhesion and cell migration. Pituitary adenomas are a common human health problem, yet most cases are sporadic, necessitating alternative approaches to traditional Mendelian genetic studies. Mouse models of adenoma formation offer the opportunity for gene expression profiling during progressive stages of hyperplasia, adenoma and tumorigenesis. This approach holds promise for the identification of relevant pathways and candidate genes as risk factors for adenoma formation, understanding mechanisms of progression, and identifying drug targets and clinically relevant biomarkers.


Subject(s)
Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Pituitary Gland/metabolism , Pituitary Neoplasms/genetics , Pituitary Neoplasms/metabolism , Animals , Computational Biology/methods , Disease Models, Animal , Gene Expression Profiling , Mice , Mice, Mutant Strains , Pituitary Gland/pathology , Pituitary Hormones/deficiency , Pituitary Hormones/genetics , Pituitary Hormones/metabolism , Pituitary Neoplasms/pathology
20.
Genomics ; 93(5): 449-60, 2009 May.
Article in English | MEDLINE | ID: mdl-19121383

ABSTRACT

We report a catalog of the mouse embryonic pituitary gland transcriptome consisting of five cDNA libraries including wild type tissue from E12.5 and E14.5, Prop1(df/df) mutant at E14.5, and two cDNA subtractions: E14.5 WT-E14.5 Prop1(df/df) and E14.5 WT-E12.5 WT. DNA sequence information is assembled into a searchable database with gene ontology terms representing 12,009 expressed genes. We validated coverage of the libraries by detecting most known homeobox gene transcription factor cDNAs. A total of 45 homeobox genes were detected as part of the pituitary transcriptome, representing most expected ones, which validated library coverage, and many novel ones, underscoring the utility of this resource as a discovery tool. We took a similar approach for signaling-pathway members with novel pituitary expression and found 157 genes related to the BMP, FGF, WNT, SHH and NOTCH pathways. These genes are exciting candidates for regulators of pituitary development and function.


Subject(s)
Computational Biology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Pituitary Gland/embryology , Signal Transduction/genetics , Transcription Factors/metabolism , Animals , Bone Morphogenetic Protein Receptors, Type I/genetics , Gene Library , Genomics , Homeodomain Proteins/genetics , Mice , Models, Animal , Pituitary Gland/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...