1
|
Gordon SD, Duffy DL, Whiteman DC, Olsen CM, McAloney K, Adsett JM, Garden NA, Cross SM, List-Armitage SE, Brown J, Beck JJ, Mbarek H, Medland SE, Montgomery GW, Martin NG. GWAS of Dizygotic Twinning in an Enlarged Australian Sample of Mothers of DZ Twins. Twin Res Hum Genet 2023:1-12. [PMID: 37994447 DOI: 10.1017/thg.2023.45] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Female fertility is a complex trait with age-specific changes in spontaneous dizygotic (DZ) twinning and fertility. To elucidate factors regulating female fertility and infertility, we conducted a genome-wide association study (GWAS) on mothers of spontaneous DZ twins (MoDZT) versus controls (3273 cases, 24,009 controls). This is a follow-up study to the Australia/New Zealand (ANZ) component of that previously reported (Mbarek et al., 2016), with a sample size almost twice that of the entire discovery sample meta-analysed in the previous article (and five times the ANZ contribution to that), resulting from newly available additional genotyping and representing a significant increase in power. We compare analyses with and without male controls and show unequivocally that it is better to include male controls who have been screened for recent family history, than to use only female controls. Results from the SNP based GWAS identified four genomewide significant signals, including one novel region, ZFPM1 (Zinc Finger Protein, FOG Family Member 1), on chromosome 16. Previous signals near FSHB (Follicle Stimulating Hormone beta subunit) and SMAD3 (SMAD Family Member 3) were also replicated (Mbarek et al., 2016). We also ran the GWAS with a dominance model that identified a further locus ADRB2 on chr 5. These results have been contributed to the International Twinning Genetics Consortium for inclusion in the next GWAS meta-analysis (Mbarek et al., in press).
Collapse
Affiliation(s)
- Scott D Gordon
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - David L Duffy
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - David C Whiteman
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Catherine M Olsen
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Kerrie McAloney
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Jessica M Adsett
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Natalie A Garden
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Simone M Cross
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | | | - Joy Brown
- Independent researcher, Invercargill, New Zealand
| | - Jeffrey J Beck
- Avera Institute for Human Genetics, Avera McKennan Hospital and University Health Center, Sioux Falls, South Dakota, USA
| | | | - Sarah E Medland
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Grant W Montgomery
- Institute of Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Nicholas G Martin
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| |
Collapse
|
2
|
Daly AZ, Mortensen AH, Bando H, Camper SA. Pituitary Tumors and Immortalized Cell Lines Generated by Cre-Inducible Expression of SV40 T Antigen. Endocrinology 2021; 162:6219492. [PMID: 33837405 PMCID: PMC8183496 DOI: 10.1210/endocr/bqab073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Indexed: 02/07/2023]
Abstract
Targeted oncogenesis is the process of driving tumor formation by engineering transgenic mice that express an oncogene under the control of a cell-type specific promoter. Such tumors can be adapted to cell culture, providing immortalized cell lines. To make it feasible to follow the process of tumorigenesis and increase the opportunity for generating cell lines, we developed a mouse strain that expresses SV40 T antigens in response to Cre-recombinase. Using CRISPR/Cas9 we inserted a cassette with coding sequences for SV40 T antigens and an internal ribosome entry site with green fluorescent protein cassette (IRES-GFP) into the Rosa26 locus, downstream from a stop sequence flanked by loxP sites: Rosa26LSL-SV40-GFP. These mice were mated with previously established Prop1-cre and Tshb-cre transgenic lines. Both the Rosa26LSL-SV40-GFP/+; Prop1-cre and Rosa26LSL-SV40-GFP/+; Tshb-cre mice developed fully penetrant dwarfism and large tumors by 4 weeks. Tumors from both of these mouse lines were adapted to growth in cell culture. We have established a progenitor-like cell line (PIT-P1) that expresses Sox2 and Pitx1, and a thyrotrope-like cell line (PIT-T1) that expresses Pou1f1 and Cga. These studies demonstrate the utility of the novel, Rosa26LSL-SV40-GFP mouse line for reliable targeted oncogenesis and development of unique cell lines.
Collapse
Affiliation(s)
| | | | - Hironori Bando
- University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Sally A Camper
- University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Correspondence: Sally A. Camper, Ph.D., 5704 Medical Science Building II, 1301 Catherine St, Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
3
|
Ryan GE, Bohaczuk SC, Cassin J, Witham EA, Shojaei S, Ho EV, Thackray VG, Mellon PL. Androgen receptor positively regulates gonadotropin-releasing hormone receptor in pituitary gonadotropes. Mol Cell Endocrinol 2021; 530:111286. [PMID: 33872733 PMCID: PMC8177864 DOI: 10.1016/j.mce.2021.111286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 03/29/2021] [Accepted: 04/13/2021] [Indexed: 11/30/2022]
Abstract
Within pituitary gonadotropes, the gonadotropin-releasing hormone receptor (GnRHR) receives hypothalamic input from GnRH neurons that is critical for reproduction. Previous studies have suggested that androgens may regulate GnRHR, although the mechanisms remain unknown. In this study, we demonstrated that androgens positively regulate Gnrhr mRNA in mice. We then investigated the effects of androgens and androgen receptor (AR) on Gnrhr promoter activity in immortalized mouse LβT2 cells, which represent mature gonadotropes. We found that AR positively regulates the Gnrhr proximal promoter, and that this effect requires a hormone response element (HRE) half site at -159/-153 relative to the transcription start site. We also identified nonconsensus, full-length HREs at -499/-484 and -159/-144, which are both positively regulated by androgens on a heterologous promoter. Furthermore, AR associates with the Gnrhr promoter in ChIP. Altogether, we report that GnRHR is positively regulated by androgens through recruitment of AR to the Gnrhr proximal promoter.
Collapse
Affiliation(s)
- Genevieve E Ryan
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
| | - Stephanie C Bohaczuk
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
| | - Jessica Cassin
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
| | - Emily A Witham
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
| | - Shadi Shojaei
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
| | - Emily V Ho
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
| | - Varykina G Thackray
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
| | - Pamela L Mellon
- Department of Obstetrics, Gynecology and Reproductive Sciences, Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
4
|
Xie H, Hoffmann HM, Iyer AK, Brayman MJ, Ngo C, Sunshine MJ, Mellon PL. Chromatin status and transcription factor binding to gonadotropin promoters in gonadotrope cell lines. Reprod Biol Endocrinol 2017; 15:86. [PMID: 29065928 PMCID: PMC5655979 DOI: 10.1186/s12958-017-0304-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 10/04/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Proper expression of key reproductive hormones from gonadotrope cells of the pituitary is required for pubertal onset and reproduction. To further our understanding of the molecular events taking place during embryonic development, leading to expression of the glycoproteins luteinizing hormone (LH) and follicle-stimulating hormone (FSH), we characterized chromatin structure changes, imparted mainly by histone modifications, in model gonadotrope cell lines. METHODS We evaluated chromatin status and gene expression profiles by chromatin immunoprecipitation assays, DNase sensitivity assay, and RNA sequencing in three developmentally staged gonadotrope cell lines, αT1-1 (progenitor, expressing Cga), αT3-1 (immature, expressing Cga and Gnrhr), and LβT2 (mature, expressing Cga, Gnrhr, Lhb, and Fshb), to assess changes in chromatin status and transcription factor access of gonadotrope-specific genes. RESULTS We found the common mRNA α-subunit of LH and FSH, called Cga, to have an open chromatin conformation in all three cell lines. In contrast, chromatin status of Gnrhr is open only in αT3-1 and LβT2 cells. Lhb begins to open in LβT2 cells and was further opened by activin treatment. Histone H3 modifications associated with active chromatin were high on Gnrhr in αT3-1 and LβT2, and Lhb in LβT2 cells, while H3 modifications associated with repressed chromatin were low on Gnrhr, Lhb, and Fshb in LβT2 cells. Finally, chromatin status correlates with the progressive access of LHX3 to Cga and Gnrhr, followed by PITX1 binding to the Lhb promoter. CONCLUSION Our data show the gonadotrope-specific genes Cga, Gnrhr, Lhb, and Fshb are not only controlled by developmental transcription factors, but also by epigenetic mechanisms that include the modulation of chromatin structure, and histone modifications.
Collapse
Affiliation(s)
- Huimin Xie
- 0000 0001 2107 4242grid.266100.3Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California, 9500 Gilman Drive La Jolla, San Diego, CA 92093-0674 USA
| | - Hanne M. Hoffmann
- 0000 0001 2107 4242grid.266100.3Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California, 9500 Gilman Drive La Jolla, San Diego, CA 92093-0674 USA
| | - Anita K. Iyer
- 0000 0001 2107 4242grid.266100.3Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California, 9500 Gilman Drive La Jolla, San Diego, CA 92093-0674 USA
- 0000 0004 0507 3954grid.185669.5Illumina Inc, 5200 Illumina Way, San Diego, CA 92122 USA
| | - Melissa J. Brayman
- 0000 0001 2107 4242grid.266100.3Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California, 9500 Gilman Drive La Jolla, San Diego, CA 92093-0674 USA
- Foley and Lardner LLP, 402 West Broadway, Suite 2100, San Diego, CA 92101 USA
| | - Cindy Ngo
- 0000 0001 2107 4242grid.266100.3Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California, 9500 Gilman Drive La Jolla, San Diego, CA 92093-0674 USA
| | - Mary Jean Sunshine
- 0000 0001 2107 4242grid.266100.3Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California, 9500 Gilman Drive La Jolla, San Diego, CA 92093-0674 USA
| | - Pamela L. Mellon
- 0000 0001 2107 4242grid.266100.3Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California, 9500 Gilman Drive La Jolla, San Diego, CA 92093-0674 USA
| |
Collapse
|
5
|
Laverrière JN, L'Hôte D, Tabouy L, Schang AL, Quérat B, Cohen-Tannoudji J. Epigenetic regulation of alternative promoters and enhancers in progenitor, immature, and mature gonadotrope cell lines. Mol Cell Endocrinol 2016; 434:250-65. [PMID: 27402603 DOI: 10.1016/j.mce.2016.07.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 07/05/2016] [Accepted: 07/05/2016] [Indexed: 11/25/2022]
Abstract
Gonadotrope cell identity genes emerge in a stepwise process during mouse pituitary development. Cga, encoding for the α-subunit of TSH, LH, and FSH, is initially detected at E11.5 followed by Gnrhr and steroidogenic factor Sf1 at E13.5, specifying cells engaged in a gonadotrope cell fate. Lhb and Fshb appear at E16.5 and 17.5, respectively, typifying differentiated gonadotrope cells. Using the αT1-1, αT3-1 and LβT2 cell lines recapitulating these stages of gonadotrope differentiation, DNA methylation at Gnrhr and Sf1 was investigated. Regulatory regions were found hypermethylated in progenitor αT1-1 cells and hypomethylated in differentiated LβT2 cells. Abundance of RNA polymerase II together with active histone modifications including H3K4me1, H3K4me3, and H3K27ac were strictly correlated with DNA hypomethylation. Analyses of epigenomic modifications and chromatin accessibility were further extended to Isl1, Lhx3, Gata2, and Pitx2, highlighting alternative usages of specific regulatory gene domains in progenitor αT1-1, immature αT3-1, and mature LβT2 gonadotrope cells.
Collapse
Affiliation(s)
- Jean-Noël Laverrière
- Univ Paris Diderot, Sorbonne Paris Cité, Biologie Fonctionnelle et Adaptative (BFA), F-75013, Paris, France; CNRS UMR 8251, F-75013, Paris, France; Physiologie de l'axe gonadotrope INSERM U1133, F-75013, Paris, France.
| | - David L'Hôte
- Univ Paris Diderot, Sorbonne Paris Cité, Biologie Fonctionnelle et Adaptative (BFA), F-75013, Paris, France; CNRS UMR 8251, F-75013, Paris, France; Physiologie de l'axe gonadotrope INSERM U1133, F-75013, Paris, France
| | - Laure Tabouy
- Univ Paris Diderot, Sorbonne Paris Cité, Biologie Fonctionnelle et Adaptative (BFA), F-75013, Paris, France; CNRS UMR 8251, F-75013, Paris, France; Physiologie de l'axe gonadotrope INSERM U1133, F-75013, Paris, France
| | - Anne-Laure Schang
- Univ Paris Diderot, Sorbonne Paris Cité, Biologie Fonctionnelle et Adaptative (BFA), F-75013, Paris, France; CNRS UMR 8251, F-75013, Paris, France; Physiologie de l'axe gonadotrope INSERM U1133, F-75013, Paris, France
| | - Bruno Quérat
- Univ Paris Diderot, Sorbonne Paris Cité, Biologie Fonctionnelle et Adaptative (BFA), F-75013, Paris, France; CNRS UMR 8251, F-75013, Paris, France; Physiologie de l'axe gonadotrope INSERM U1133, F-75013, Paris, France
| | - Joëlle Cohen-Tannoudji
- Univ Paris Diderot, Sorbonne Paris Cité, Biologie Fonctionnelle et Adaptative (BFA), F-75013, Paris, France; CNRS UMR 8251, F-75013, Paris, France; Physiologie de l'axe gonadotrope INSERM U1133, F-75013, Paris, France
| |
Collapse
|
6
|
Hadziselimovic F, Gegenschatz-Schmid K, Verkauskas G, Docampo-Garcia MJ, Demougin P, Bilius V, Malcius D, Dasevicius D, Stadtler MB. Gene Expression Changes Underlying Idiopathic Central Hypogonadism in Cryptorchidism with Defective Mini-Puberty. Sex Dev 2016; 10:136-46. [PMID: 27561106 DOI: 10.1159/000447762] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2016] [Indexed: 11/19/2022] Open
Abstract
The whole genome RNA profiling of testicular biopsies by DNA strand-specific RNA sequencing was examined to determine a potential causative role of isolated congenital cryptorchidism in azoospermia and/or infertility in the context of our previously published GeneChip data. Cryptorchid patients, aged 7 months to 5 years and otherwise healthy, were enrolled in this prospective study. During surgery, testicular tissue biopsies were obtained for histological examination and RNA sequencing. Fifteen patients were selected based on the histological results and were divided into 2 groups. Seven were classified as belonging to the high infertility risk (HIR) and 8 to the low infertility risk (LIR) group. Cryptorchid boys in the HIR group lacked transformation of gonocytes into Ad spermatogonia due to impaired mini-puberty. This group of patients will be infertile despite successful surgery. The new important finding was a decreased PROK2, CHD7, FGFR1, and SPRY4 gene expression in the HIR group. Furthermore, identification of multiple differences in gene expression between HIR and LIR groups underscores the importance of an intact hypothalamic-pituitary-gonadal axis for fertility development. Our RNA profiling data strongly support the theory that in the HIR group of cryptorchid boys insufficient PROK2/CHD7/FGFR1/SPRY4 gene expression induces deficient LH secretion, resulting in impaired mini-puberty and infertility. We therefore recommend hormonal treatment for this cohort of cryptorchid boys with defective mini-puberty following a seemingly successful orchidopexy.
Collapse
|
7
|
McDonald EA, Smith JE, Cederberg RA, White BR. Divergent activity of the gonadotropin-releasing hormone receptor gene promoter among genetic lines of pigs is partially conferred by nuclear factor (NF)-B, specificity protein (SP)1-like and GATA-4 binding sites. Reprod Biol Endocrinol 2016; 14:36. [PMID: 27356969 PMCID: PMC4928339 DOI: 10.1186/s12958-016-0170-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 06/22/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Binding of gonadotropin-releasing hormone (GnRH) to its receptor (GnRHR) on gonadotropes within the anterior pituitary gland is essential to reproduction. In pigs, the GnRHR gene is also located near a genetic marker for ovulation rate, a primary determinant of prolificacy. We hypothesized that pituitary expression of the GnRHR gene is alternatively regulated in genetic strains with elevated ovulation rates (Chinese Meishan and Nebraska Index) vs. standard white crossbred swine (Control). METHODS Luciferase reporter vectors containing 5118 bp of GnRHR gene promoter from either the Control, Index or Meishan swine lines were generated. Transient transfection of line-specific, full length, deletion and mutation constructs into gonadotrope-derived αT3-1 cells were performed to compare promoter activity and identify regions necessary for divergent regulation of the porcine GnRHR gene. Additionally, transcription factors that bind the GnRHR promoter from each line were identified with electrophoretic mobility shift assays (EMSA). RESULTS Dramatic differences in luciferase activity among Control, Index and Meishan promoters (19-, 27- and 49-fold over promoterless control, respectively; P < 0.05) were established. A single bp substitution (-1690) within a previously identified upstream enhancer (-1779/-1667) bound GATA-4 in the Meishan promoter and the p52/p65 subunits of nuclear factor (NF)-κB in the homologous Control/Index promoters. Transient transfection of vectors containing block replacement mutations of either the GATA-4 or NF-κB binding sites within the context of their native promoters resulted in a 50 and 60 % reduction of luciferase activity, respectively (P < 0.05). Furthermore, two single-bp substitutions in the Meishan compared to Control/Index promoters resulted in binding of the p52 and p65 subunits of NF-κB and a specificity protein 1 (SP1)-like factor (-1235) as well as GATA-4 (-845). Vectors containing the full-length Meishan promoter harboring individual mutations spanning these regions reduced luciferase activity by 25 and 20 %, respectively, compared to native sequence (P < 0.05). CONCLUSIONS Elevated activity of the Meishan GnRHR gene promoter over Control/Index promoters in αT3-1 cells is partially due to three single nucleotide polymorphisms resulting in the unique binding of GATA-4 (-1690), the p52/p65 subunits of NF-kB in combination with a SP1-like factor (-1235), and GATA-4 (-845).
Collapse
Affiliation(s)
- Emily A. McDonald
- Laboratory of Reproductive Biology, Department of Animal Science, Institute of Agriculture and Natural Resources, University of Nebraska-Lincoln, Lincoln, NE USA
- Present address: Center for International Health Research, Rhode Island Hospital, Providence, RI USA
| | - Jacqueline E. Smith
- Laboratory of Reproductive Biology, Department of Animal Science, Institute of Agriculture and Natural Resources, University of Nebraska-Lincoln, Lincoln, NE USA
- Present address: Stowers Institute for Medical Research, Kansas City, MO USA
| | - Rebecca A. Cederberg
- Laboratory of Reproductive Biology, Department of Animal Science, Institute of Agriculture and Natural Resources, University of Nebraska-Lincoln, Lincoln, NE USA
| | - Brett R. White
- Laboratory of Reproductive Biology, Department of Animal Science, Institute of Agriculture and Natural Resources, University of Nebraska-Lincoln, Lincoln, NE USA
| |
Collapse
|
8
|
Brauer VM, Wiarda-Bell JR, Desaulniers AT, Cederberg RA, White BR. Functional activity of the porcine Gnrhr2 gene promoter in testis-derived cells is partially conferred by nuclear factor-κB, specificity protein 1 and 3 (SP1/3) and overlapping early growth response 1/SP1/3 binding sites. Gene 2016; 587:137-46. [PMID: 27134031 DOI: 10.1016/j.gene.2016.04.052] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 04/02/2016] [Accepted: 04/26/2016] [Indexed: 11/30/2022]
Abstract
Unlike the classical gonadotropin-releasing hormone (GnRH1), the second mammalian isoform (GnRH2) is ubiquitously expressed, suggesting a divergent function. Indeed, we demonstrated that GnRH2 governs LH-independent testosterone secretion in porcine testes via interaction with its receptor (GnRHR2) on Leydig cells. Transient transfections with luciferase reporter vectors containing 3009bp of 5' flanking sequence for the porcine Gnrhr2 gene (-3009pGL3) revealed promoter activity in all 15 cell lines examined, including swine testis-derived (ST) cells. Therefore, ST cells were utilized to explore the molecular mechanisms underlying transcriptional regulation of the porcine Gnrhr2 gene in the testis. Reporter plasmids containing progressive 5' deletions of the Gnrhr2 promoter indicated that the -708/-490 region contained elements critical to promoter activity. Electrophoretic mobility shift assays (EMSAs) with radiolabeled oligonucleotides spanning the -708/-490bp region and ST nuclear extracts, identified specific binding complexes for the -513/-490, -591/-571 and -606/-581bp segments of promoter. Antibody addition to EMSAs indicated that the p65 and p52 subunits of nuclear factor-κB (NF-κB) comprised the specific complex bound to the oligonucleotide probe for the -513/-490bp promoter region, specificity protein (SP) 1 and 3 bound the -591/-571bp probe and early growth response 1 (EGR1), SP1 and SP3 bound the -606/-581 radiolabeled oligonucleotide. Transient transfections with vectors containing mutations of the NF-κB (-499/-493), SP1/3 (-582/-575) or overlapping EGR1/SP1/3 (-597/-587) binding sites reduced luciferase activity by 26%, 61% and 56%, respectively (P<0.05). Thus, NF-κB, SP1/3 and overlapping EGR1/SP1/3 binding sites are critical to expression of the porcine Gnrhr2 gene in ST cells.
Collapse
Affiliation(s)
- Vanessa M Brauer
- Laboratory of Reproductive Biology, Department of Animal Science, Institute of Agriculture and Natural Resources, University of Nebraska-Lincoln, Lincoln, NE 68583-0908, USA
| | - Jocelyn R Wiarda-Bell
- Laboratory of Reproductive Biology, Department of Animal Science, Institute of Agriculture and Natural Resources, University of Nebraska-Lincoln, Lincoln, NE 68583-0908, USA
| | - Amy T Desaulniers
- Laboratory of Reproductive Biology, Department of Animal Science, Institute of Agriculture and Natural Resources, University of Nebraska-Lincoln, Lincoln, NE 68583-0908, USA
| | - Rebecca A Cederberg
- Laboratory of Reproductive Biology, Department of Animal Science, Institute of Agriculture and Natural Resources, University of Nebraska-Lincoln, Lincoln, NE 68583-0908, USA
| | - Brett R White
- Laboratory of Reproductive Biology, Department of Animal Science, Institute of Agriculture and Natural Resources, University of Nebraska-Lincoln, Lincoln, NE 68583-0908, USA.
| |
Collapse
|
9
|
Wang H, Hastings R, Miller WL, Kumar TR. Fshb-iCre mice are efficient and specific Cre deleters for the gonadotrope lineage. Mol Cell Endocrinol 2016; 419:124-38. [PMID: 26472536 PMCID: PMC4684453 DOI: 10.1016/j.mce.2015.10.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 10/06/2015] [Accepted: 10/07/2015] [Indexed: 02/06/2023]
Abstract
Genetic analysis of development and function of the gonadotrope cell lineage within mouse anterior pituitary has been greatly facilitated by at least three currently available Cre strains in which Cre was either knocked into the Gnrhr locus or expressed as a transgene from Cga and Lhb promoters. However, in each case there are some limitations including CRE expression in thyrotropes within pituitary or ectopic expression outside of pituitary, for example in some populations of neurons or gonads. Hence, these Cre strains often pose problems with regard to undesirable deletion of alleles in non-gonadotrope cells, fertility and germline transmission of mutant alleles. Here, we describe generation and characterization of a new Fshb-iCre deleter strain using 4.7 kb of ovine Fshb promoter regulatory sequences driving iCre expression exclusively in the gonadotrope lineage within anterior pituitary. Fshb-iCre mice develop normally, display no ectopic CRE expression in gonads and are fertile. When crossed onto a loxP recombination-mediated red to green color switch reporter mouse genetic background, in vivo CRE recombinase activity is detectable in gonadotropes at more than 95% efficiency and the GFP-tagged gonadotropes readily purified by fluorescence activated cell sorting. We demonstrate the applicability of this Fshb-iCre deleter strain in a mouse model in which Dicer is efficiently and selectively deleted in gonadotropes. We further show that loss of DICER-dependent miRNAs in gonadotropes leads to profound suppression of gonadotropins resulting in male and female infertility. Thus, Fshb-iCre mice serve as a new genetic tool to efficiently manipulate gonadotrope-specific gene expression in vivo.
Collapse
Affiliation(s)
- Huizhen Wang
- Department of Molecular and Integrative Physiology
| | | | - William L Miller
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, NC, 27695, USA
| | - T Rajendra Kumar
- Department of Molecular and Integrative Physiology; Center for Reproductive Sciences, Institute for Reproductive Health and Regenerative Medicine; Department of Pathology and Laboratory Medicine; Department of Neurosurgery, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| |
Collapse
|
10
|
Xie H, Hoffmann HM, Meadows JD, Mayo SL, Trang C, Leming SS, Maruggi C, Davis SW, Larder R, Mellon PL. Homeodomain Proteins SIX3 and SIX6 Regulate Gonadotrope-specific Genes During Pituitary Development. Mol Endocrinol 2015; 29:842-55. [PMID: 25915183 PMCID: PMC4447639 DOI: 10.1210/me.2014-1279] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 04/20/2015] [Indexed: 12/15/2022] Open
Abstract
Sine oculis-related homeobox 3 (SIX3) and SIX6, 2 closely related homeodomain transcription factors, are involved in development of the mammalian neuroendocrine system and mutations of Six6 adversely affect fertility in mice. We show that both small interfering RNA knockdown in gonadotrope cell lines and knockout of Six6 in both embryonic and adult male mice (Six6 knockout) support roles for SIX3 and SIX6 in transcriptional regulation in gonadotrope gene expression and that SIX3 and SIX6 can functionally compensate for each other. Six3 and Six6 expression patterns in gonadotrope cell lines reflect the timing of the expression of pituitary markers they regulate. Six3 is expressed in an immature gonadotrope cell line and represses transcription of the early lineage-specific pituitary genes, GnRH receptor (GnRHR) and the common α-subunit (Cga), whereas Six6 is expressed in a mature gonadotrope cell line and represses the specific β-subunits of LH and FSH (LHb and FSHb) that are expressed later in development. We show that SIX6 repression requires interaction with transducin-like enhancer of split corepressor proteins and competition for DNA-binding sites with the transcriptional activator pituitary homeobox 1. Our studies also suggest that estradiol and circadian rhythm regulate pituitary expression of Six6 and Six3 in adult females but not in males. In summary, SIX3 and SIX6 play distinct but compensatory roles in regulating transcription of gonadotrope-specific genes as gonadotrope cells differentiate.
Collapse
Affiliation(s)
- Huimin Xie
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (H.X., H.M.H., J.D.M., S.L.M., C.T., S.S.L., C.M., R.L., P.L.M.), University of California, San Diego, La Jolla, California 92093; and Department of Human Genetics (S.W.D.), University of Michigan, Ann Arbor, Michigan 48109
| | - Hanne M Hoffmann
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (H.X., H.M.H., J.D.M., S.L.M., C.T., S.S.L., C.M., R.L., P.L.M.), University of California, San Diego, La Jolla, California 92093; and Department of Human Genetics (S.W.D.), University of Michigan, Ann Arbor, Michigan 48109
| | - Jason D Meadows
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (H.X., H.M.H., J.D.M., S.L.M., C.T., S.S.L., C.M., R.L., P.L.M.), University of California, San Diego, La Jolla, California 92093; and Department of Human Genetics (S.W.D.), University of Michigan, Ann Arbor, Michigan 48109
| | - Susan L Mayo
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (H.X., H.M.H., J.D.M., S.L.M., C.T., S.S.L., C.M., R.L., P.L.M.), University of California, San Diego, La Jolla, California 92093; and Department of Human Genetics (S.W.D.), University of Michigan, Ann Arbor, Michigan 48109
| | - Crystal Trang
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (H.X., H.M.H., J.D.M., S.L.M., C.T., S.S.L., C.M., R.L., P.L.M.), University of California, San Diego, La Jolla, California 92093; and Department of Human Genetics (S.W.D.), University of Michigan, Ann Arbor, Michigan 48109
| | - Sunamita S Leming
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (H.X., H.M.H., J.D.M., S.L.M., C.T., S.S.L., C.M., R.L., P.L.M.), University of California, San Diego, La Jolla, California 92093; and Department of Human Genetics (S.W.D.), University of Michigan, Ann Arbor, Michigan 48109
| | - Chiara Maruggi
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (H.X., H.M.H., J.D.M., S.L.M., C.T., S.S.L., C.M., R.L., P.L.M.), University of California, San Diego, La Jolla, California 92093; and Department of Human Genetics (S.W.D.), University of Michigan, Ann Arbor, Michigan 48109
| | - Shannon W Davis
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (H.X., H.M.H., J.D.M., S.L.M., C.T., S.S.L., C.M., R.L., P.L.M.), University of California, San Diego, La Jolla, California 92093; and Department of Human Genetics (S.W.D.), University of Michigan, Ann Arbor, Michigan 48109
| | - Rachel Larder
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (H.X., H.M.H., J.D.M., S.L.M., C.T., S.S.L., C.M., R.L., P.L.M.), University of California, San Diego, La Jolla, California 92093; and Department of Human Genetics (S.W.D.), University of Michigan, Ann Arbor, Michigan 48109
| | - Pamela L Mellon
- Department of Reproductive Medicine and the Center for Reproductive Science and Medicine (H.X., H.M.H., J.D.M., S.L.M., C.T., S.S.L., C.M., R.L., P.L.M.), University of California, San Diego, La Jolla, California 92093; and Department of Human Genetics (S.W.D.), University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
11
|
Cederberg RA, Smith JE, McDonald EA, Lee C, Perkins AR, White BR. Activity of the porcine gonadotropin-releasing hormone receptor gene promoter is partially conferred by a distal gonadotrope specific element (GSE) within an upstream enhancing region, two proximal GSEs and a retinoid X receptor binding site. Reprod Biol Endocrinol 2015; 13:45. [PMID: 25981521 PMCID: PMC4461931 DOI: 10.1186/s12958-015-0033-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 04/16/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Regulation of gonadotropin-releasing hormone (GnRH) receptor (GnRHR) numbers on gonadotropes within the anterior pituitary gland represents a critical point for control of reproductive function. Binding of GnRH to its receptor regulates follicle stimulating hormone (FSH) and luteinizing hormone (LH) release and levels of this G-protein coupled receptor on the surface of gonadotropes determines their sensitivity to GnRH pulses. While transcriptional regulation of this gene has been studied in mice, rats, humans and sheep, little is known about its regulation in the pig, an important agricultural species and human research model. METHODS We isolated 5118 bp of 5' flanking sequence for the porcine GnRHR gene and generated luciferase reporter vectors. Deletion and mutation constructs were evaluated in gonadotrope-derived alphaT3-1 cells to determine regions important for gene transcription. Additionally, electrophoretic mobility shift assays (EMSAs) were performed to identify transcription factors binding to the GnRHR promoter. RESULTS Transient transfections revealed that the GnRHR promoter was functional in alphaT3-1 cells but not in cells of non-gonadotrope origin. Mutation of the highly conserved gonadotrope specific element (GSE) located at -179/-171 of proximal promoter completely ablated luciferase activity, whereas mutation of another GSE at -315/-310 reduced activity by 34%. Consistent with this, EMSAs using alphaT3-1 nuclear extracts and a steroidogenic factor (SF)1 antibody confirmed SF1 binding to both GSEs. EMSAs also demonstrated that a retinoid X receptor (RXR) binding site at -279/-274 binds RXRalpha and RXRbeta and mutation of this site eliminated promoter activity. Transient transfection of alphaT3-1 cells with reporter vectors containing selective removal of 5' flanking region for the porcine GnRHR gene indicated that the -1915/-1431 segment was important for promoter activity. Definition of this region via transfection assays and EMSAs revealed an upstream enhancing region located at -1779/-1667 that increases porcine GnRHR gene expression in alphaT3-1 cells and includes a SF1 binding site at -1760/-1753. CONCLUSIONS Porcine GnRHR promoter activity in alphaT3-1 cells is partially conferred by a distal GSE, two proximal GSEs and a RXR binding site. Basal gonadotrope expression of the porcine GnRHR gene uniquely involves three GSEs and RXR is newly identified as a regulator of GnRHR promoter activity.
Collapse
Affiliation(s)
- Rebecca A Cederberg
- Laboratory of Reproductive Biology, Department of Animal Science, Institute of Agriculture and Natural Resources, University of Nebraska-Lincoln, Lincoln, Nebraska, USA.
| | - Jacqueline E Smith
- Laboratory of Reproductive Biology, Department of Animal Science, Institute of Agriculture and Natural Resources, University of Nebraska-Lincoln, Lincoln, Nebraska, USA.
- Current address: Stowers Institute for Medical Research, Kansas City, MO, USA.
| | - Emily A McDonald
- Laboratory of Reproductive Biology, Department of Animal Science, Institute of Agriculture and Natural Resources, University of Nebraska-Lincoln, Lincoln, Nebraska, USA.
- Current address: Center for International Health Research, Rhode Island Hospital, Providence, RI, USA.
| | - Chanho Lee
- Laboratory of Reproductive Biology, Department of Animal Science, Institute of Agriculture and Natural Resources, University of Nebraska-Lincoln, Lincoln, Nebraska, USA.
| | - Amy R Perkins
- Laboratory of Reproductive Biology, Department of Animal Science, Institute of Agriculture and Natural Resources, University of Nebraska-Lincoln, Lincoln, Nebraska, USA.
- Current address: Arizona Andrology Laboratory and Cryobank, Tuscon, AZ, USA.
| | - Brett R White
- Laboratory of Reproductive Biology, Department of Animal Science, Institute of Agriculture and Natural Resources, University of Nebraska-Lincoln, Lincoln, Nebraska, USA.
| |
Collapse
|
12
|
Fortin J, Ongaro L, Li Y, Tran S, Lamba P, Wang Y, Zhou X, Bernard DJ. Minireview: Activin Signaling in Gonadotropes: What Does the FOX say… to the SMAD? Mol Endocrinol 2015; 29:963-77. [PMID: 25942106 DOI: 10.1210/me.2015-1004] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The activins were discovered and named based on their abilities to stimulate FSH secretion and FSHβ (Fshb) subunit expression by pituitary gonadotrope cells. According to subsequent in vitro observations, activins also stimulate the transcription of the GnRH receptor (Gnrhr) and the activin antagonist, follistatin (Fst). Thus, not only do activins stimulate FSH directly, they have the potential to regulate both FSH and LH indirectly by modulating gonadotrope sensitivity to hypothalamic GnRH. Moreover, activins may negatively regulate their own actions by stimulating the production of one of their principal antagonists. Here, we describe our current understanding of the mechanisms through which activins regulate Fshb, Gnrhr, and Fst transcription in vitro. The activin signaling molecules SMAD3 and SMAD4 appear to partner with the winged-helix/forkhead transcription factor, forkhead box L2 (FOXL2), to regulate expression of all 3 genes. However, in vivo data paint a different picture. Although conditional deletion of Foxl2 and/or Smad4 in murine gonadotropes produces impairments in FSH synthesis and secretion as well as in pituitary Fst expression, Gnrhr mRNA levels are either unperturbed or increased in these animals. Surprisingly, gonadotrope-specific deletion of Smad3 alone or with Smad2 does not impair FSH production or fertility; however, mice harboring these mutations may express a DNA binding-deficient, but otherwise functional, SMAD3 protein. Collectively, the available data firmly establish roles for FOXL2 and SMAD4 in Fshb and Fst expression in gonadotrope cells, whereas SMAD3's role requires further investigation. Gnrhr expression, in contrast, appears to be FOXL2, SMAD4, and, perhaps, activin independent in vivo.
Collapse
Affiliation(s)
- Jérôme Fortin
- Department of Pharmacology and Therapeutics (J.F., L.O., Y.L., S.T., P.L., Y.W., X.Z., D.J.B.), McGill University, Montréal, Québec, Canada H3G 1Y6; The Campbell Family Cancer Research Institute (J.F.), Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada M5G 2C1; Diabetes Center (S.T.), Department of Medicine, University of California-San Francisco, San Francisco, California 94143; and Psychiatry (P.L.), St Mary Mercy Hospital, Livonia, Michigan 48154
| | - Luisina Ongaro
- Department of Pharmacology and Therapeutics (J.F., L.O., Y.L., S.T., P.L., Y.W., X.Z., D.J.B.), McGill University, Montréal, Québec, Canada H3G 1Y6; The Campbell Family Cancer Research Institute (J.F.), Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada M5G 2C1; Diabetes Center (S.T.), Department of Medicine, University of California-San Francisco, San Francisco, California 94143; and Psychiatry (P.L.), St Mary Mercy Hospital, Livonia, Michigan 48154
| | - Yining Li
- Department of Pharmacology and Therapeutics (J.F., L.O., Y.L., S.T., P.L., Y.W., X.Z., D.J.B.), McGill University, Montréal, Québec, Canada H3G 1Y6; The Campbell Family Cancer Research Institute (J.F.), Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada M5G 2C1; Diabetes Center (S.T.), Department of Medicine, University of California-San Francisco, San Francisco, California 94143; and Psychiatry (P.L.), St Mary Mercy Hospital, Livonia, Michigan 48154
| | - Stella Tran
- Department of Pharmacology and Therapeutics (J.F., L.O., Y.L., S.T., P.L., Y.W., X.Z., D.J.B.), McGill University, Montréal, Québec, Canada H3G 1Y6; The Campbell Family Cancer Research Institute (J.F.), Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada M5G 2C1; Diabetes Center (S.T.), Department of Medicine, University of California-San Francisco, San Francisco, California 94143; and Psychiatry (P.L.), St Mary Mercy Hospital, Livonia, Michigan 48154
| | - Pankaj Lamba
- Department of Pharmacology and Therapeutics (J.F., L.O., Y.L., S.T., P.L., Y.W., X.Z., D.J.B.), McGill University, Montréal, Québec, Canada H3G 1Y6; The Campbell Family Cancer Research Institute (J.F.), Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada M5G 2C1; Diabetes Center (S.T.), Department of Medicine, University of California-San Francisco, San Francisco, California 94143; and Psychiatry (P.L.), St Mary Mercy Hospital, Livonia, Michigan 48154
| | - Ying Wang
- Department of Pharmacology and Therapeutics (J.F., L.O., Y.L., S.T., P.L., Y.W., X.Z., D.J.B.), McGill University, Montréal, Québec, Canada H3G 1Y6; The Campbell Family Cancer Research Institute (J.F.), Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada M5G 2C1; Diabetes Center (S.T.), Department of Medicine, University of California-San Francisco, San Francisco, California 94143; and Psychiatry (P.L.), St Mary Mercy Hospital, Livonia, Michigan 48154
| | - Xiang Zhou
- Department of Pharmacology and Therapeutics (J.F., L.O., Y.L., S.T., P.L., Y.W., X.Z., D.J.B.), McGill University, Montréal, Québec, Canada H3G 1Y6; The Campbell Family Cancer Research Institute (J.F.), Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada M5G 2C1; Diabetes Center (S.T.), Department of Medicine, University of California-San Francisco, San Francisco, California 94143; and Psychiatry (P.L.), St Mary Mercy Hospital, Livonia, Michigan 48154
| | - Daniel J Bernard
- Department of Pharmacology and Therapeutics (J.F., L.O., Y.L., S.T., P.L., Y.W., X.Z., D.J.B.), McGill University, Montréal, Québec, Canada H3G 1Y6; The Campbell Family Cancer Research Institute (J.F.), Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada M5G 2C1; Diabetes Center (S.T.), Department of Medicine, University of California-San Francisco, San Francisco, California 94143; and Psychiatry (P.L.), St Mary Mercy Hospital, Livonia, Michigan 48154
| |
Collapse
|
13
|
Schang A. Inside and outside the pituitary: comparative analysis of Gnrhr expression provides insight into the mechanisms underlying the evolution of gene expression. J Neuroendocrinol 2015; 27:177-86. [PMID: 25556311 DOI: 10.1111/jne.12253] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Revised: 11/18/2014] [Accepted: 12/27/2014] [Indexed: 02/06/2023]
Abstract
DNA cis-acting elements involved in gene regulation may actively contribute to adaptation processes because they are submitted to lower evolutionary constraints than coding DNA. In this regard, comparisons of the mechanisms underlying basal and regulated Gnrhr expression have revealed some features that promote stable and consistent Gnrhr expression in pituitary gonadotroph cells in different species. The presence of two divergent SF1 (NR5A1) response elements in all analysed mammalian Gnrhr promoters probably comprises one of the features that ensures reliable expression in the pituitary. By contrast, in other tissues, such as the hippocampus and testis, our analyses revealed dissimilar levels of Gnrhr expression among species. Indeed, Gnrhr was consistently expressed after birth in the rat but not the mouse hippocampus. Similar discrepancies were observed in foetal and adult testes. The ability of the rat promoter to drive reporter gene expression in the hippocampus and testis of transgenic mice just as it naturally directs the expression of the endogenous Gnrhr in rats strongly suggests that regulatory DNA sequences contained species-specific instructions prevailing over other controls. The major conclusion emerging from these studies is that Gnrhr promoter sequences are mainly responsible for directing transcriptional programmes and play a predominant role over the species-specific cell environment.
Collapse
Affiliation(s)
- Al Schang
- Sorbonne Paris Cité, Biologie Fonctionnelle et Adaptative, Université Paris Diderot, Paris 7, CNRS 8251, Paris, France; Sorbonne Paris Cité, Epigénétique et Destin Cellulaire, Universite Paris Diderot, Paris 7, CNRS 7216, Paris, France
| |
Collapse
|
14
|
Castinetti F, Reynaud R, Quentien MH, Jullien N, Marquant E, Rochette C, Herman JP, Saveanu A, Barlier A, Enjalbert A, Brue T. Combined pituitary hormone deficiency: current and future status. J Endocrinol Invest 2015; 38:1-12. [PMID: 25200994 DOI: 10.1007/s40618-014-0141-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 07/17/2014] [Indexed: 12/20/2022]
Abstract
Over the last two decades, the understanding of the mechanisms involved in pituitary ontogenesis has largely increased. Since the first description of POU1F1 human mutations responsible for a well-defined phenotype without extra-pituitary malformation, several other genetic defects of transcription factors have been reported with variable degrees of phenotype-genotype correlations. However, to date, despite the identification of an increased number of genetic causes of isolated or multiple pituitary deficiencies, the etiology of most (80-90 %) congenital cases of hypopituitarism remains unsolved. Identifying new etiologies is of importance as a post-natal diagnosis to better diagnose and treat the patients (delayed pituitary deficiencies, differential diagnosis of a pituitary mass on MRI, etc.), and as a prenatal diagnosis to decrease the risk of early death (undiagnosed corticotroph deficiency for instance). The aim of this review is to summarize the main etiologies and phenotypes of combined pituitary hormone deficiencies, associated or not with extra-pituitary anomalies, and to suggest how the identification of such etiologies could be improved in the near future.
Collapse
Affiliation(s)
- F Castinetti
- Aix-Marseille Université, CNRS, Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille CRN2M UMR 7286, cedex 15, 13344, Marseille, France.
- APHM, Hôpital Timone Adultes, Service d'Endocrinologie, Diabète et Maladies Métaboliques, cedex 5, 13385, Marseille, France.
- Centre de Référence des Maladies Rares d'Origine Hypophysaire DEFHY, cedex 15, 13385, Marseille, France.
| | - R Reynaud
- Aix-Marseille Université, CNRS, Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille CRN2M UMR 7286, cedex 15, 13344, Marseille, France
- APHM, Hôpital Timone Enfants, Service de Pédiatrie multidisciplinaire, cedex 5, 13385, Marseille, France
- Centre de Référence des Maladies Rares d'Origine Hypophysaire DEFHY, cedex 15, 13385, Marseille, France
| | - M-H Quentien
- Aix-Marseille Université, CNRS, Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille CRN2M UMR 7286, cedex 15, 13344, Marseille, France
- APHM, Hôpital Timone Adultes, Service d'Endocrinologie, Diabète et Maladies Métaboliques, cedex 5, 13385, Marseille, France
- Centre de Référence des Maladies Rares d'Origine Hypophysaire DEFHY, cedex 15, 13385, Marseille, France
| | - N Jullien
- Aix-Marseille Université, CNRS, Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille CRN2M UMR 7286, cedex 15, 13344, Marseille, France
| | - E Marquant
- Aix-Marseille Université, CNRS, Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille CRN2M UMR 7286, cedex 15, 13344, Marseille, France
- APHM, Hôpital Timone Enfants, Service de Pédiatrie multidisciplinaire, cedex 5, 13385, Marseille, France
- Centre de Référence des Maladies Rares d'Origine Hypophysaire DEFHY, cedex 15, 13385, Marseille, France
| | - C Rochette
- Aix-Marseille Université, CNRS, Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille CRN2M UMR 7286, cedex 15, 13344, Marseille, France
- APHM, Hôpital Timone Adultes, Service d'Endocrinologie, Diabète et Maladies Métaboliques, cedex 5, 13385, Marseille, France
- Centre de Référence des Maladies Rares d'Origine Hypophysaire DEFHY, cedex 15, 13385, Marseille, France
| | - J-P Herman
- Aix-Marseille Université, CNRS, Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille CRN2M UMR 7286, cedex 15, 13344, Marseille, France
| | - A Saveanu
- Aix-Marseille Université, CNRS, Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille CRN2M UMR 7286, cedex 15, 13344, Marseille, France
- APHM, Hôpital Timone Adultes, Service d'Endocrinologie, Diabète et Maladies Métaboliques, cedex 5, 13385, Marseille, France
- APHM, Hôpital de la Conception, Laboratoire de Biologie Moléculaire, 13005, Marseille, France
- Centre de Référence des Maladies Rares d'Origine Hypophysaire DEFHY, cedex 15, 13385, Marseille, France
| | - A Barlier
- Aix-Marseille Université, CNRS, Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille CRN2M UMR 7286, cedex 15, 13344, Marseille, France
- APHM, Hôpital Timone Adultes, Service d'Endocrinologie, Diabète et Maladies Métaboliques, cedex 5, 13385, Marseille, France
- APHM, Hôpital de la Conception, Laboratoire de Biologie Moléculaire, 13005, Marseille, France
- Centre de Référence des Maladies Rares d'Origine Hypophysaire DEFHY, cedex 15, 13385, Marseille, France
| | - A Enjalbert
- Aix-Marseille Université, CNRS, Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille CRN2M UMR 7286, cedex 15, 13344, Marseille, France
- APHM, Hôpital de la Conception, Laboratoire de Biologie Moléculaire, 13005, Marseille, France
- Centre de Référence des Maladies Rares d'Origine Hypophysaire DEFHY, cedex 15, 13385, Marseille, France
| | - T Brue
- Aix-Marseille Université, CNRS, Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille CRN2M UMR 7286, cedex 15, 13344, Marseille, France
- APHM, Hôpital Timone Adultes, Service d'Endocrinologie, Diabète et Maladies Métaboliques, cedex 5, 13385, Marseille, France
- Centre de Référence des Maladies Rares d'Origine Hypophysaire DEFHY, cedex 15, 13385, Marseille, France
| |
Collapse
|
15
|
Park S, Mullen RD, Rhodes SJ. Cell-specific actions of a human LHX3 gene enhancer during pituitary and spinal cord development. Mol Endocrinol 2013; 27:2013-27. [PMID: 24100213 DOI: 10.1210/me.2013-1161] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
The LIM class of homeodomain protein 3 (LHX3) transcription factor is essential for pituitary gland and nervous system development in mammals. In humans, mutations in the LHX3 gene underlie complex pediatric syndromes featuring deficits in anterior pituitary hormones and defects in the nervous system. The mechanisms that control temporal and spatial expression of the LHX3 gene are poorly understood. The proximal promoters of the human LHX3 gene are insufficient to guide expression in vivo and downstream elements including a conserved enhancer region appear to play a role in tissue-specific expression in the pituitary and nervous system. Here we characterized the activity of this downstream enhancer region in regulating gene expression at the cellular level during development. Human LHX3 enhancer-driven Cre reporter transgenic mice were generated to facilitate studies of enhancer actions. The downstream LHX3 enhancer primarily guides gene transcription in α-glycoprotein subunit -expressing cells secreting the TSHβ, LHβ, or FSHβ hormones and expressing the GATA2 and steroidogenic factor 1 transcription factors. In the developing nervous system, the enhancer serves as a targeting module active in V2a interneurons. These results demonstrate that the downstream LHX3 enhancer is important in specific endocrine and neural cell types but also indicate that additional regulatory elements are likely involved in LHX3 gene expression. Furthermore, these studies revealed significant gonadotrope cell heterogeneity during pituitary development, providing insights into the cellular physiology of this key reproductive regulatory cell. The human LHX3 enhancer-driven Cre reporter transgenic mice also provide a valuable tool for further developmental studies of cell determination and differentiation in the pituitary and nervous system.
Collapse
Affiliation(s)
- Soyoung Park
- PhD, Department of Biology, Indiana University-Purdue University Indianapolis, LD222, 402 North Blackford Street, Indianapolis, IN 46202-5120.
| | | | | |
Collapse
|
16
|
Larder R, Kimura I, Meadows J, Clark DD, Mayo S, Mellon PL. Gene dosage of Otx2 is important for fertility in male mice. Mol Cell Endocrinol 2013; 377:16-22. [PMID: 23811236 PMCID: PMC3771655 DOI: 10.1016/j.mce.2013.06.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 06/18/2013] [Accepted: 06/19/2013] [Indexed: 02/07/2023]
Abstract
Together, the hypothalamus, pituitary and gonads direct the development and regulation of reproductive function in mammals. Gonadotropin-releasing hormone (GnRH) expression is limited to ∼800 neurons that originate in the olfactory placode then migrate to the hypothalamus. Coordination of the hypothalamic-pituitary-gonadal (HPG) axis is dependent upon correct neuronal migration of GnRH neurons into the hypothalamus followed by proper synthesis and pulsatile secretion of GnRH. Defects in any one of these processes causes infertility. Otx2, the vertebrate homologue of Drosophila orthodenticle, is a transcription factor that has been shown to be critical for normal brain and eye development and is expressed in both the developing GnRH neurons and the pituitary, suggesting that this gene may play a critical role in development of the HPG axis. As Otx2-null mice are embryonic lethal, we have analyzed the reproductive capacity of heterozygous Otx2 mice to determine the contribution of Otx2 gene dosage to normal HPG axis function. Our data reveal that correct dosage of Otx2 is critical for normal fertility as loss of one allele of Otx2 leads to a discernible reproductive phenotype in male mice due to disruption of the migration of GnRH neurons during development.
Collapse
Affiliation(s)
- Rachel Larder
- Department of Reproductive Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0674
| | - Ikuo Kimura
- Department of Reproductive Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0674
- Department of Genomic Drug Discovery Science, Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo-ku, Kyoto 606-8501, Japan
| | - Jason Meadows
- Department of Reproductive Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0674
| | - Daniel. D. Clark
- Department of Reproductive Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0674
| | - Susan Mayo
- Department of Reproductive Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0674
| | - Pamela L. Mellon
- Department of Reproductive Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0674
- To whom correspondence should be addressed, , Telephone: 1-858-534-1312, Fax: 1-858-534-1438
| |
Collapse
|
17
|
Hunter CS, Malik RE, Witzmann FA, Rhodes SJ. LHX3 interacts with inhibitor of histone acetyltransferase complex subunits LANP and TAF-1β to modulate pituitary gene regulation. PLoS One 2013; 8:e68898. [PMID: 23861948 PMCID: PMC3701669 DOI: 10.1371/journal.pone.0068898] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 06/07/2013] [Indexed: 01/19/2023] Open
Abstract
LIM-homeodomain 3 (LHX3) is a transcription factor required for mammalian pituitary gland and nervous system development. Human patients and animal models with LHX3 gene mutations present with severe pediatric syndromes that feature hormone deficiencies and symptoms associated with nervous system dysfunction. The carboxyl terminus of the LHX3 protein is required for pituitary gene regulation, but the mechanism by which this domain operates is unknown. In order to better understand LHX3-dependent pituitary hormone gene transcription, we used biochemical and mass spectrometry approaches to identify and characterize proteins that interact with the LHX3 carboxyl terminus. This approach identified the LANP/pp32 and TAF-1β/SET proteins, which are components of the inhibitor of histone acetyltransferase (INHAT) multi-subunit complex that serves as a multifunctional repressor to inhibit histone acetylation and modulate chromatin structure. The protein domains of LANP and TAF-1β that interact with LHX3 were mapped using biochemical techniques. Chromatin immunoprecipitation experiments demonstrated that LANP and TAF-1β are associated with LHX3 target genes in pituitary cells, and experimental alterations of LANP and TAF-1β levels affected LHX3-mediated pituitary gene regulation. Together, these data suggest that transcriptional regulation of pituitary genes by LHX3 involves regulated interactions with the INHAT complex.
Collapse
Affiliation(s)
- Chad S. Hunter
- Department of Biology, Indiana University-Purdue University Indianapolis, Indiana, United States of America
| | - Raleigh E. Malik
- Department of Biochemistry and Molecular Biology, Indiana School of Medicine, Indianapolis, Indiana, United States of America
| | - Frank A. Witzmann
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Simon J. Rhodes
- Department of Biology, Indiana University-Purdue University Indianapolis, Indiana, United States of America
- Department of Biochemistry and Molecular Biology, Indiana School of Medicine, Indianapolis, Indiana, United States of America
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- * E-mail:
| |
Collapse
|
18
|
Prince KL, Colvin SC, Park S, Lai X, Witzmann FA, Rhodes SJ. Developmental analysis and influence of genetic background on the Lhx3 W227ter mouse model of combined pituitary hormone deficiency disease. Endocrinology 2013; 154:738-48. [PMID: 23288907 PMCID: PMC3548188 DOI: 10.1210/en.2012-1790] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Combined pituitary hormone deficiency (CPHD) diseases result in severe outcomes for patients including short stature, developmental delays, and reproductive deficiencies. Little is known about their etiology, especially the developmental profiles and the influences of genetic background on disease progression. Animal models for CPHD provide valuable tools to investigate disease mechanisms and inform diagnostic and treatment protocols. Here we examined hormone production during pituitary development and the influence of genetic background on phenotypic severity in the Lhx3(W227ter/W227ter) mouse model. Lhx3(W227ter/W227ter) embryos have deficiencies of ACTH, α-glycoprotein subunit, GH, PRL, TSHβ, and LHβ during prenatal development. Furthermore, mutant mice have significant reduction in the critical pituitary transcriptional activator-1 (PIT1). Through breeding, the Lhx3(W227ter/W227ter) genotype was placed onto the 129/Sv and C57BL/6 backgrounds. Intriguingly, the genetic background significantly affected viability: whereas Lhx3(W227ter/W227ter) animals were found in the expected frequencies in C57BL/6, homozygous animals were not viable in the 129/Sv genetic environment. The hormone marker and PIT1 reductions observed in Lhx3(W227ter/W227ter) mice on a mixed background were also seen in the separate strains but in some cases were more severe in 129/Sv. To further characterize the molecular changes in diseased mice, we conducted a quantitative proteomic analysis of pituitary proteins. This showed significantly lower levels of PRL, pro-opiomelanocortin (ACTH), and α-glycoprotein subunit proteins in Lhx3(W227ter/W227ter) mice. Together, these data show that hormone deficiency disease is apparent in early prenatal stages in this CPHD model system. Furthermore, as is noted in human disease, genetic background significantly impacts the phenotypic outcome of these monogenic endocrine diseases.
Collapse
Affiliation(s)
- Kelly L Prince
- Departments of Cellular and Integrative Physiology, Indiana University-Purdue University, Indianapolis, IN 46202, USA
| | | | | | | | | | | |
Collapse
|
19
|
Xie H, Cherrington BD, Meadows JD, Witham EA, Mellon PL. Msx1 homeodomain protein represses the αGSU and GnRH receptor genes during gonadotrope development. Mol Endocrinol 2013; 27:422-36. [PMID: 23371388 DOI: 10.1210/me.2012-1289] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Multiple homeodomain transcription factors are crucial for pituitary organogenesis and cellular differentiation. A homeodomain repressor, Msx1, is expressed from the ventral aspect of the developing anterior pituitary and implicated in gonadotrope differentiation. Here, we find that Msx1 represses transcription of lineage-specific pituitary genes such as the common α-glycoprotein subunit (αGSU) and GnRH receptor (GnRHR) promoters in the mouse gonadotrope-derived cell lines, αT3-1 and LβT2. Repression of the mouse GnRHR promoter by Msx1 is mediated through a consensus-binding motif in the downstream activin regulatory element (DARE). Truncation and mutation analyses of the human αGSU promoter map Msx1 repression to a site at -114, located at the junctional regulatory element (JRE). Dlx activators are closely related to the Msx repressors, acting through the same elements, and Dlx3 and Dlx2 act as transcriptional activators for GnRHR and αGSU, respectively. Small interfering RNA knockdown of Msx1 in αT3-1 cells increases endogenous αGSU and GnRHR mRNA expression. Msx1 gene expression reaches its maximal expression at the rostral edge at e13.5. The subsequent decline in Msx1 expression specifically coincides with the onset of expression of both αGSU and GnRHR. The expression levels of both αGSU and GnRHR in Msx1-null mice at e18.5 are higher compared with wild type, further confirming a role for Msx1 in the repression of αGSU and GnRHR. In summary, Msx1 functions as a negative regulator early in pituitary development by repressing the gonadotrope-specific αGSU and GnRHR genes, but a temporal decline in Msx1 expression alleviates this repression allowing induction of GnRHR and αGSU, thus serving to time the onset of gonadotrope-specific gene program.
Collapse
Affiliation(s)
- Huimin Xie
- Department of Reproductive Medicine, Center for Reproductive Science and Medicine, University of California San Diego, La Jolla, CA 92093-0674, USA
| | | | | | | | | |
Collapse
|
20
|
Schang AL, Bleux C, Chenut MC, Ngô-Muller V, Quérat B, Jeanny JC, Counis R, Cohen-Tannoudji J, Laverrière JN. Identification and analysis of two novel sites of rat GnRH receptor gene promoter activity: the pineal gland and retina. Neuroendocrinology 2013; 97:115-31. [PMID: 22414758 DOI: 10.1159/000337661] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 02/28/2012] [Indexed: 11/19/2022]
Abstract
BACKGROUND AND AIMS In mammals, activation of pituitary GnRH receptor (GnRHR) by hypothalamic GnRH increases the synthesis and secretion of LH and FSH, which, in turn, regulate gonadal functions. However, GnRHR gene (Gnrhr) expression is not restricted to the pituitary. METHODS To gain insight into the extrapituitary expression of Gnrhr, a transgenic mouse model that expresses the human placental alkaline phosphatase reporter gene driven by the rat Gnrhr promoter was created. RESULTS This study shows that the rat Gnrhr promoter is operative in two functionally related organs, the pineal gland, as early as embryonic day (E) 13.5, and the retina where activity was only detected at E17.5. Accordingly, Gnrhr mRNA were present in both tissues. Transcription factors known to regulate Gnrhr promoter activity such as the LIM homeodomain factors LHX3 and ISL1 were also detected in the retina. Furthermore, transient transfection studies in CHO and gonadotrope cells revealed that OTX2, a major transcription factor in both pineal and retina cell differentiation, is able to activate the Gnrhr promoter together with either CREB or PROP1, depending on the cell context. CONCLUSION Rather than using alternate promoters, Gnrhr expression is directed to diverse cell lineages through specific associations of transcription factors acting on distinct response elements along the same promoter. These data open new avenues regarding GnRH-mediated control of seasonal and circadian rhythms in reproductive physiology.
Collapse
Affiliation(s)
- Anne-Laure Schang
- Université Paris Diderot Paris 7, Sorbonne Paris Cité, Biologie Fonctionnelle et Adaptative, EAC CNRS 4413, Physiologie de l'Axe Gonadotrope, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Schang AL, Granger A, Quérat B, Bleux C, Cohen-Tannoudji J, Laverrière JN. GATA2-induced silencing and LIM-homeodomain protein-induced activation are mediated by a bi-functional response element in the rat GnRH receptor gene. Mol Endocrinol 2012; 27:74-91. [PMID: 23211524 DOI: 10.1210/me.2012-1182] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
GATA2 transcription factor and LIM homeodomain proteins Islet1 (ISL1) and LIM homeobox 3 (LHX3) are suspected to be involved in gonadotrope cell fate and maintenance. The GnRH receptor gene (Gnrhr), crucial for gonadotrope function, is expressed in the pituitary gland from embryonic day 13.5 onward, well before LH and FSH β-subunits. This expression pattern together with the presence of WGATAR and TAAT motifs in Gnrhr promoter sequences suggests the involvement of early transcription factors in promoter activation. In this study, using a well-characterized transgenic mouse model, GATA2 was found colocalized with Gnrhr promoter activity in the pituitary. Transient transfection of Gnrhr promoter luciferase fusion constructs together with either GATA2 expression vectors or small interfering RNA in gonadotrope cell lines indicated that GATA2, which typically acts as a trans-activator, unexpectedly repressed Gnrhr promoter activity. Using DNA chromatography affinity and EMSA, we demonstrated that GATA2 operates via a response element containing a peculiar palindromic GATA motif that overlaps a critical TAAT motif involved in LHX3/ISL1 trans-activation. Indeed, despite the inhibitory action of GATA2, this element displayed a clear-cut enhancer activity in gonadotrope cells. Chromatin immunoprecipitation assays indicated that GATA2, LHX3, and ISL1 interact with a Gnrhr promoter fragment encompassing this element. The trans-repressive action of GATA2 on Gnrhr promoter activity is likely balanced or even hindered by trans-activating effects of LIM homeodomain proteins via this novel bifunctional LIM/GATA response element. Such a hierarchical interplay may contribute to finely adjust Gnrhr gene expression in gonadotrope cell lineage during pituitary development as well as in the adult animal.
Collapse
Affiliation(s)
- Anne-Laure Schang
- University of Paris Diderot Paris 7, Sorbonne Paris Cité, Biologie Fonctionnelle et Adaptative, Centre National de la Recherche Scientifique Equipe d'Accueil Conventionnée 4413, Physiologie de l'Axe Gonadotrope, Bâtiment Buffon, Case Courrier 7007, 75205 Paris Cedex 13, France
| | | | | | | | | | | |
Collapse
|
22
|
Schang AL, Quérat B, Simon V, Garrel G, Bleux C, Counis R, Cohen-Tannoudji J, Laverrière JN. Mechanisms underlying the tissue-specific and regulated activity of the Gnrhr promoter in mammals. Front Endocrinol (Lausanne) 2012; 3:162. [PMID: 23248618 PMCID: PMC3521148 DOI: 10.3389/fendo.2012.00162] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 11/28/2012] [Indexed: 01/27/2023] Open
Abstract
The GnRH receptor (GnRHR) plays a central role in the development and maintenance of reproductive function in mammals. Following stimulation by GnRH originating from the hypothalamus, GnRHR triggers multiple signaling events that ultimately stimulate the synthesis and the periodic release of the gonadotropins, luteinizing-stimulating hormone (LH) and follicle-stimulating hormones (FSH) which, in turn, regulate gonadal functions including steroidogenesis and gametogenesis. The concentration of GnRHR at the cell surface is essential for the amplitude and the specificity of gonadotrope responsiveness. The number of GnRHR is submitted to strong regulatory control during pituitary development, estrous cycle, pregnancy, lactation, or after gonadectomy. These modulations take place, at least in part, at the transcriptional level. To analyze this facet of the reproductive function, the 5' regulatory sequences of the gene encoding the GnRHR have been isolated and characterized through in vitro and in vivo approaches. This review summarizes results obtained with the mouse, rat, human, and ovine promoters either by transient transfection assays or by means of transgenic mice.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jean-Noël Laverrière
- *Correspondence: Jean-Noël Laverrière, Physiologie de l’Axe Gonadotrope, Biologie Fonctionnelle et Adaptative, EAC CNRS 4413, Sorbonne Paris Cité, Université Paris Diderot-Paris 7, Bâtiment Buffon, case courrier 7007, 4 rue MA Lagroua Weill-Hallé, 75205 Paris Cedex 13, France. e-mail:
| |
Collapse
|
23
|
Liu JB, Lan XY, Xu Y, Li ZJ, Lei CZ, Chen H. Combined effects of three novel SNPs within goat LHX3 gene on milk performance. Genes Genomics 2011. [DOI: 10.1007/s13258-011-0026-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
24
|
Prince KL, Walvoord EC, Rhodes SJ. The role of homeodomain transcription factors in heritable pituitary disease. Nat Rev Endocrinol 2011; 7:727-37. [PMID: 21788968 DOI: 10.1038/nrendo.2011.119] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The anterior pituitary gland secretes hormones that regulate developmental and physiological processes, including growth, the stress response, metabolic status, reproduction and lactation. During embryogenesis, cellular determination and differentiation events establish specialized hormone-secreting cell types within the anterior pituitary gland. These developmental decisions are mediated in part by the actions of a cascade of transcription factors, many of which belong to the homeodomain class of DNA-binding proteins. The discovery of some of these regulatory proteins has facilitated genetic analyses of patients with hormone deficiencies. The findings of these studies reveal that congenital defects-ranging from isolated hormone deficiencies to combined pituitary hormone deficiency syndromes-are sometimes associated with mutations in the genes encoding pituitary-acting developmental transcription factors. The phenotypes of affected individuals and animal models have together provided useful insights into the biology of these transcription factors and have suggested new hypotheses for testing in the basic science laboratory. Here, we summarize the gene regulatory pathways that control anterior pituitary development, with emphasis on the role of the homeodomain transcription factors in normal pituitary organogenesis and heritable pituitary disease.
Collapse
Affiliation(s)
- Kelly L Prince
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Medical Science Room 362A, 635 North Barnhill Drive, Indianapolis, IN 46202-5120, USA
| | | | | |
Collapse
|
25
|
Abstract
The hypothalamus, pituitary, and gonads coordinate to direct the development and regulation of reproductive function in mammals. Control of the hypothalamic-pituitary-gonadal axis is dependent on correct migration of gonadotropin-releasing hormone (GnRH) neurons from the nasal placode to the hypothalamus, followed by proper synthesis and pulsatile secretion of GnRH, functions absent in patients with hypogonadal hypogonadism. In this study, we identify sine oculis-related homeobox 6 (Six6) as a novel factor necessary for proper targeting of GnRH expression to the limited population of GnRH neurons within the adult mouse hypothalamus and demonstrate that it is required for proper reproductive function in both male and female mice. Female Six6-null mice exhibit a striking decrease in fertility, failing to progress through the estrous cycle normally, show any signs of successful ovulation, or produce litters. Although basal gonadotropin production in these mice is relatively normal, analysis of GnRH expression reveals a dramatic decrease in total GnRH neuron numbers. We show that expression of Six6 is dramatically increased during GnRH neuronal maturation and that overexpression of Six6 induces GnRH transcription in neuronal cells. Finally, we demonstrate that this induction in GnRH expression is mediated via binding of Six6 to evolutionarily conserved ATTA sites located within the GnRH proximal promoter. Together, these data indicate that Six6 plays an important role in the regulation of GnRH expression and hypothalamic control of fertility.
Collapse
|
26
|
Model of pediatric pituitary hormone deficiency separates the endocrine and neural functions of the LHX3 transcription factor in vivo. Proc Natl Acad Sci U S A 2010; 108:173-8. [PMID: 21149718 DOI: 10.1073/pnas.1009501108] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The etiology of most pediatric hormone deficiency diseases is poorly understood. Children with combined pituitary hormone deficiency (CPHD) have insufficient levels of multiple anterior pituitary hormones causing short stature, metabolic disease, pubertal failure, and often have associated nervous system symptoms. Mutations in developmental regulatory genes required for the specification of the hormone-secreting cell types of the pituitary gland underlie severe forms of CPHD. To better understand these diseases, we have created a unique mouse model of CPHD with a targeted knockin mutation (Lhx3 W227ter), which is a model for the human LHX3 W224ter disease. The LHX3 gene encodes a LIM-homeodomain transcription factor, which has essential roles in pituitary and nervous system development in mammals. The introduced premature termination codon results in deletion of the carboxyl terminal region of the LHX3 protein, which is critical for pituitary gene activation. Mice that lack all LHX3 function do not survive beyond birth. By contrast, the homozygous Lhx3 W227ter mice survive, but display marked dwarfism, thyroid disease, and female infertility. Importantly, the Lhx3 W227ter mice have no apparent nervous system deficits. The Lhx3 W227ter mouse model provides a unique array of hormone deficits and facilitates experimental approaches that are not feasible with human patients. These experiments demonstrate that the carboxyl terminus of the LHX3 transcription factor is not required for viability. More broadly, this study reveals that the in vivo actions of a transcription factor in different tissues are molecularly separable.
Collapse
|
27
|
Breen KM, Thackray VG, Coss D, Mellon PL. Runt-related transcription factors impair activin induction of the follicle-stimulating hormone {beta}-subunit gene. Endocrinology 2010; 151:2669-80. [PMID: 20357224 PMCID: PMC2875819 DOI: 10.1210/en.2009-0949] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Synthesis of the FSH beta-subunit (FSHbeta) is critical for normal reproduction in mammals, and its expression within the pituitary gonadotrope is tightly regulated by activin. Here we show that Runt-related (RUNX) proteins, transcriptional regulators known to interact with TGFbeta signaling pathways, suppress activin induction of FSHbeta gene expression. Runx2 is expressed within the murine pituitary gland and dramatically represses activin-induced FSHbeta promoter activity, without affecting basal expression in LbetaT2 cells, an immortalized mouse gonadotrope cell line. This repressive effect is specific, because RUNX2 induces LHbeta transcription (with or without activin) and does not interfere with GnRH induction of either gonadotropin beta-subunit gene. Analysis of the murine FSHbeta promoter by transfection and gel shift assays reveals that RUNX2 repression localizes to a Runx-binding element at -159/-153, which is adjacent to a previously recognized region critical for activin induction. Mutation of this -153 activin-response element or, indeed, any of the five activin-responsive regions prevents activin induction and, in fact, RUNX2 suppression, instead converting RUNX2 to an activator of the FSHbeta gene. Although the Runx-binding element is required for RUNX2-mediated repression of FSHbeta induction by either activin or Smad3, confirming a functional role of this novel site, protein interactions in addition to those between RUNX2 and Smads are necessary to account for full repression of activin induction. In summary, the present study provides evidence for Runx2-mediated repression of activin-induced FSHbeta gene expression and reveals the context dependence of Runx2 action in hormonal regulation of the gonadotropin genes.
Collapse
Affiliation(s)
- Kellie M Breen
- Department of Reproductive Medicine and Center for Reproductive Science and Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0674, USA
| | | | | | | |
Collapse
|
28
|
Kelberman D, Rizzoti K, Lovell-Badge R, Robinson ICAF, Dattani MT. Genetic regulation of pituitary gland development in human and mouse. Endocr Rev 2009; 30:790-829. [PMID: 19837867 PMCID: PMC2806371 DOI: 10.1210/er.2009-0008] [Citation(s) in RCA: 268] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Normal hypothalamopituitary development is closely related to that of the forebrain and is dependent upon a complex genetic cascade of transcription factors and signaling molecules that may be either intrinsic or extrinsic to the developing Rathke's pouch. These factors dictate organ commitment, cell differentiation, and cell proliferation within the anterior pituitary. Abnormalities in these processes are associated with congenital hypopituitarism, a spectrum of disorders that includes syndromic disorders such as septo-optic dysplasia, combined pituitary hormone deficiencies, and isolated hormone deficiencies, of which the commonest is GH deficiency. The highly variable clinical phenotypes can now in part be explained due to research performed over the last 20 yr, based mainly on naturally occurring and transgenic animal models. Mutations in genes encoding both signaling molecules and transcription factors have been implicated in the etiology of hypopituitarism, with or without other syndromic features, in mice and humans. To date, mutations in known genes account for a small proportion of cases of hypopituitarism in humans. However, these mutations have led to a greater understanding of the genetic interactions that lead to normal pituitary development. This review attempts to describe the complexity of pituitary development in the rodent, with particular emphasis on those factors that, when mutated, are associated with hypopituitarism in humans.
Collapse
Affiliation(s)
- Daniel Kelberman
- Developmental Endocrinology Research Group, Clinical and Molecular Genetics Unit, Institute of Child Health, 30 Guilford Street, London WC1N 1EH, United Kingdom
| | | | | | | | | |
Collapse
|
29
|
Lents CA, Farmerie TA, Cherrington BD, Clay CM. Multiple core homeodomain binding motifs differentially contribute to transcriptional activity of the murine gonadotropin-releasing hormone receptor gene promoter. Endocrine 2009; 35:356-64. [PMID: 19333792 DOI: 10.1007/s12020-009-9167-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2008] [Revised: 01/21/2009] [Accepted: 02/23/2009] [Indexed: 11/25/2022]
Abstract
Multiple homeodomain (Hbox) proteins have been shown to organize expression of key markers of gonadotropes. Nine putative Hbox-binding sites, characterized by the homeospecific TAAT motif, are located within the proximal 600 bp of the murine GnRHR promoter. Homeoproteins bind separate Hbox sites within this promoter, supporting basal- and endocrine-directed transcription. The function of the most proximal sites (Hbox1 and Hbox2) in the murine GnRHR is unknown; thus, understanding of the global contribution of homeospecific TAAT sites to promoter function is incomplete. Site-directed mutagenesis revealed that loss of Hbox2 reduced promoter activity in a cell-specific manner, having no effect in alphaT3-1 cells but reducing promoter function in LbetaT2 cells, another gonadotrope-derived cell line representing a later developmental stage. In contrast, eliminating Hbox1 reduced basal activity in both lines. This region displayed specific binding to homeoprotein Oct-1. Mutagenesis of a previously identified Oct-1-binding site in concert with Hbox1 led to further reduction in activity. We suggest that the two most proximal homeodomain-binding sites in the murine GnRHR promoter may regulate the promoter in a developmentally dependent fashion and that Oct-1 acts at multiple but distinct TAAT sites to support basal transcription.
Collapse
Affiliation(s)
- Clay A Lents
- Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, The University of Georgia, 316 Rhodes Center ADS, Athens, GA 30602, USA
| | | | | | | |
Collapse
|
30
|
Larder R, Mellon PL. Otx2 induction of the gonadotropin-releasing hormone promoter is modulated by direct interactions with Grg co-repressors. J Biol Chem 2009; 284:16966-16978. [PMID: 19401468 PMCID: PMC2719334 DOI: 10.1074/jbc.m109.002485] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hormonal communication between the hypothalamus, pituitary, and gonads orchestrates the development and regulation of mammalian reproductive function. In mice, gonadotropin-releasing hormone (GnRH) expression is limited to approximately 1000 neurons that originate in the olfactory placode then migrate to specific positions scattered throughout the hypothalamus. Coordination of the hypothalamic-pituitary-gonadal axis is dependent upon correct migration of GnRH neurons into the hypothalamus followed by the appropriate synthesis and pulsatile secretion of GnRH. Defects in any one of these processes can cause infertility. Recently, substantial progress has been made in identifying transcription factors, and their cofactors, that regulate not only adult expression of GnRH, but also the maturation of GnRH neurons. Here, we show that expression of Otx2, a homeodomain protein required for the formation of the forebrain, is dramatically up-regulated during GnRH neuronal maturation and that overexpression of Otx2 increases GnRH promoter activity in GnRH neuronal cell lines. Furthermore, Otx2 transcriptional activity is modulated by Grg4, a member of the Groucho-related-gene (Grg) family. Using mutational analysis, we show that a WRPW peptide motif within the Otx2 protein is required for physical interaction between Otx2 and Grg4. Without this physical interaction, Grg4 cannot repress Otx2-dependent activation of GnRH gene transcription. Taken together, these data show that Otx2 is important for GnRH expression and that direct interaction between Otx2 and Grg co-repressors regulates GnRH gene expression in hypothalamic neurons.
Collapse
Affiliation(s)
- Rachel Larder
- From the Department of Reproductive Medicine and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093-0674
| | - Pamela L Mellon
- From the Department of Reproductive Medicine and Center for Reproductive Science and Medicine, University of California, San Diego, La Jolla, California 92093-0674.
| |
Collapse
|
31
|
Jing YJ, Lan XY, Chen H, Zhang LZ, Zhang CL, Pan CY, Li MJ, Ren G, Wei TB, Zhao M. Three novel single-nucleotide polymorphisms of the bovine LHX3 gene. J Biosci 2008; 33:673-9. [DOI: 10.1007/s12038-008-0087-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
32
|
Cherrington BD, Bailey JS, Diaz AL, Mellon PL. NeuroD1 and Mash1 temporally regulate GnRH receptor gene expression in immortalized mouse gonadotrope cells. Mol Cell Endocrinol 2008; 295:106-14. [PMID: 18760324 PMCID: PMC2640340 DOI: 10.1016/j.mce.2008.07.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Revised: 07/08/2008] [Accepted: 07/25/2008] [Indexed: 01/23/2023]
Abstract
Accurate spatial and temporal expression of gonadotrope-specific genes, such as the gonadotropin-releasing hormone receptor (GnRHR) gene, is critical for gonadotrope maturation. Herein, we show that a specific E-box in the mouse GnRHR promoter binds two group A basic-helix-loop-helix (bHLH) transcription factors. Mutation of this E-box decreases expression in mouse gonadotrope-derived alphaT3-1 and LbetaT2 cell lines. Microarray and western blots show that the bHLH transcription factor NeuroD1 is strongly expressed in the gonadotrope progenitor, alphaT3-1, whereas Mash1 is strongly expressed in the more mature gonadotrope, LbetaT2. Over-expression of NeuroD1 or Mash1 increases expression of the GnRHR gene or a multimer of the E-box and this increase is lost upon mutation of the E-box. Electrophoretic mobility shift assays reveal that the GnRHR E-box binds NeuroD1 from alphaT3-1 cells, but binds Mash1 from LbetaT2 cells. The sequential binding of different members of the group A bHLH transcription factor family to mouse GnRHR E-box 3 as the gonadotrope differentiates may represent a mechanism necessary for proper spatial and temporal expression of the GnRHR during gonadotrope development.
Collapse
Affiliation(s)
| | | | | | - Pamela L. Mellon
- Corresponding Author: Pamela L. Mellon, Ph.D., Dept. of Reproductive Medicine, 0674, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0674, (858) 534-1312, Fax (858) 534-1438,
| |
Collapse
|
33
|
Castinetti F, Reynaud R, Saveanu A, Quentien MH, Albarel F, Enjalbert A, Barlier A, Brue T. Congenital pituitary hormone deficiencies: role of LHX3/LHX4 genes. Expert Rev Endocrinol Metab 2008; 3:751-760. [PMID: 30764064 DOI: 10.1586/17446651.3.6.751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
LHX3 and LHX4 are LIM domain transcription factors involved in the early steps of pituitary organogenesis. They are necessary for the proper differentiation of Rathke's pouch that gives rise to the anterior pituitary lobe. Mutations of these transcription factors are involved in congenital hypopituitarism: to date, nine mutations of LHX3 have been reported, responsible for variable pituitary hormone deficiencies and extrapituitary manifestations, including limited neck rotation. By contrast, only five LHX4 mutations have been reported, responsible for variable hormone deficiencies, and pituitary/intracranial abnormalities. Future investigations will aim to better understand human pituitary organogenesis and to shed light on the interspecies differences in the roles of these transcription factors.
Collapse
Affiliation(s)
- Frederic Castinetti
- a Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille (CRN2M), UMR6231, Faculté de Médecine Nord, Centre National de la Recherche Scientifique, Université de la Méditerranée and Centre de Référence des Déficits Hypophysaires, Hôpital de la Timone, Assistance Publique Hôpitaux de Marseille, 13385 Marseille, France
| | - Rachel Reynaud
- a Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille (CRN2M), UMR6231, Faculté de Médecine Nord, Centre National de la Recherche Scientifique, Université de la Méditerranée and Centre de Référence des Déficits Hypophysaires, Hôpital de la Timone, Assistance Publique Hôpitaux de Marseille, 13385 Marseille, France
| | - Alexandru Saveanu
- b Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille (CRN2M), UMR6231, Faculté de Médecine Nord, Centre National de la Recherche Scientifique, Université de la Méditerranée and Centre de Référence des Déficits Hypophysaires, Hôpital de la Timone, Assistance Publique Hôpitaux de Marseille, 13385 Marseille, France and Laboratoire de Biochimie-Biologie Moléculaire, Hôpital Conception, Marseille, France
| | - Marie-Helene Quentien
- a Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille (CRN2M), UMR6231, Faculté de Médecine Nord, Centre National de la Recherche Scientifique, Université de la Méditerranée and Centre de Référence des Déficits Hypophysaires, Hôpital de la Timone, Assistance Publique Hôpitaux de Marseille, 13385 Marseille, France
| | - Frederique Albarel
- a Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille (CRN2M), UMR6231, Faculté de Médecine Nord, Centre National de la Recherche Scientifique, Université de la Méditerranée and Centre de Référence des Déficits Hypophysaires, Hôpital de la Timone, Assistance Publique Hôpitaux de Marseille, 13385 Marseille, France
| | - Alain Enjalbert
- b Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille (CRN2M), UMR6231, Faculté de Médecine Nord, Centre National de la Recherche Scientifique, Université de la Méditerranée and Centre de Référence des Déficits Hypophysaires, Hôpital de la Timone, Assistance Publique Hôpitaux de Marseille, 13385 Marseille, France and Laboratoire de Biochimie-Biologie Moléculaire, Hôpital Conception, Marseille, France
| | - Anne Barlier
- b Centre de Recherche en Neurobiologie et Neurophysiologie de Marseille (CRN2M), UMR6231, Faculté de Médecine Nord, Centre National de la Recherche Scientifique, Université de la Méditerranée and Centre de Référence des Déficits Hypophysaires, Hôpital de la Timone, Assistance Publique Hôpitaux de Marseille, 13385 Marseille, France and Laboratoire de Biochimie-Biologie Moléculaire, Hôpital Conception, Marseille, France
| | - Thierry Brue
- c Centre de Recherche en neurobiologie et neurophysiologie de Marseille (CRN2M), UMR6231, Faculté de Médecine Nord, Centre National de la Recherche Scientifique, Université de la Méditerranée and Centre de Référence des déficits hypophysaires, Hôpital de la Timone, Assistance Publique Hôpitaux de Marseille, 13385 Marseille, France.
| |
Collapse
|
34
|
Tian G, Singh U, Yu Y, Ellsworth BS, Hemberger M, Geyer R, Stewart MD, Behringer RR, Fundele R. Expression and function of the LIM homeobox containing genes Lhx3 and Lhx4 in the mouse placenta. Dev Dyn 2008; 237:1517-25. [PMID: 18425848 DOI: 10.1002/dvdy.21546] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The LIM homeobox containing genes of the LIM-3 group, Lhx3 and Lhx4, are critical for normal development. Both genes are involved in the formation of the pituitary and the motoneuron system and loss of either gene causes perinatal lethality. Previous studies had shown that Lhx3 is overexpressed in hyperplastic placentas of mouse interspecies hybrids. To determine the role of LHX3 in the mouse placenta, we performed expression and function analyses. Our results show that Lhx3 exhibits specific spatial and temporal expression in the mouse placenta. However, deletion of Lhx3 does not produce a placental phenotype. To test whether this is due to functional substitution by Lhx4, we performed a phenotype analysis of Lhx3-/-; Lhx4-/- double-mutant placentas. A subset of Lhx3-/-; Lhx4-/- placentas exhibited abnormal structure of the labyrinth. However, absence of both LIM-3 genes did not interfere with placental transport nor consistently with expression of target genes such as Gnrhr. Thus, LHX3 and LHX4 appear to be dispensable for placental development and function.
Collapse
Affiliation(s)
- Geng Tian
- Department of Animal Genetics and Development, Evolutionary Biology Centre, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Blount AL, Vaughan JM, Vale WW, Bilezikjian LM. A Smad-binding Element in Intron 1 Participates in Activin-dependent Regulation of the Follistatin Gene. J Biol Chem 2008; 283:7016-26. [DOI: 10.1074/jbc.m709502200] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
|
36
|
Savage JJ, Mullen RD, Sloop KW, Colvin SC, Camper SA, Franklin CL, Rhodes SJ. Transgenic mice expressing LHX3 transcription factor isoforms in the pituitary: effects on the gonadotrope axis and sex-specific reproductive disease. J Cell Physiol 2007; 212:105-17. [PMID: 17311285 DOI: 10.1002/jcp.21010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The LHX3 transcription factor plays critical roles in pituitary and nervous system development. Mutations in the human LHX3 gene cause severe hormone deficiency diseases. The gene produces two mRNAs which can be translated to three protein isoforms. The LHX3a protein contains a central region with LIM domains and a homeodomain, and a carboxyl terminus with the major transactivation domain. LHX3b is identical to LHX3a except that it has a different amino terminus. M2-LHX3 lacks the amino terminus and LIM domains of LHX3a/b. In vitro experiments have demonstrated these three proteins have different biochemical and gene regulatory properties. Here, to investigate the effects of overexpression of LHX3 in vivo, the alpha glycoprotein subunit (alphaGSU) promoter was used to produce LHX3a, LHX3b, and M2-LHX3 in the pituitary glands of transgenic mice. Alpha GSU-beta galactosidase animals were generated as controls. Male alphaGSU-LHX3a and alphaGSU-LHX3b mice are infertile and die at a young age as a result of complications associated with obstructive uropathy including uremia. These animals have a reduced number of pituitary gonadotrope cells, low circulating gonadotropins, and possible sex hormone imbalance. Female alphaGSU-LHX3a and alphaGSU-LHX3b transgenic mice are viable but have reduced fertility. By contrast, alphaGSU-M2-LHX3 mice and control mice expressing beta galactosidase are reproductively unaffected. These overexpression studies provide insights into the properties of LHX3 during pituitary development and highlight the importance of this factor in reproductive physiology.
Collapse
Affiliation(s)
- Jesse J Savage
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Pfaeffle RW, Savage JJ, Hunter CS, Palme C, Ahlmann M, Kumar P, Bellone J, Schoenau E, Korsch E, Brämswig JH, Stobbe HM, Blum WF, Rhodes SJ. Four novel mutations of the LHX3 gene cause combined pituitary hormone deficiencies with or without limited neck rotation. J Clin Endocrinol Metab 2007; 92:1909-19. [PMID: 17327381 DOI: 10.1210/jc.2006-2177] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
CONTEXT The Lhx3 LIM-homeodomain transcription factor gene is required for development of the pituitary and motoneurons in mice. Human LHX3 gene mutations have been reported in five subjects with a phenotype consisting of GH, prolactin, TSH, LH, and FSH deficiency; abnormal pituitary morphology; and limited neck rotation. OBJECTIVE The objective of the study was to determine the frequency and nature of LHX3 mutations in patients with isolated GH deficiency or combined pituitary hormone deficiency (CPHD) and characterize the molecular consequences of mutations. DESIGN The LHX3 sequence was determined. The biochemical properties of aberrant LHX3 proteins resulting from observed mutations were characterized using reporter gene and DNA binding experiments. PATIENTS The study included 366 patients with isolated GH deficiency or CPHD. RESULTS In seven patients with CPHD from four consanguineous pedigrees, four novel, recessive mutations were identified: a deletion of the entire gene (del/del), mutations causing truncated proteins (E173ter, W224ter), and a mutation causing a substitution in the homeodomain (A210V). The mutations were associated with diminished DNA binding and pituitary gene activation, consistent with observed hormone deficiencies. Whereas subjects with del/del, E173ter, and A210V mutations had limited neck rotation, patients with the W224ter mutation did not. CONCLUSIONS LHX3 mutations are a rare cause of CPHD involving deficiencies for GH, prolactin, TSH, and LH/FSH in all patients. Whereas most patients have a severe hormone deficiency manifesting after birth, milder forms can be observed, and limited neck rotation is not a universal feature of patients with LHX3 mutations. This study extends the known molecular defects and range of phenotypes found in LHX3-associated diseases.
Collapse
Affiliation(s)
- Roland W Pfaeffle
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Medical Science Room 362A, 635 North Barnhill Drive, Indianapolis, IN 46202-5120, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
McGillivray SM, Thackray VG, Coss D, Mellon PL. Activin and glucocorticoids synergistically activate follicle-stimulating hormone beta-subunit gene expression in the immortalized LbetaT2 gonadotrope cell line. Endocrinology 2007; 148:762-73. [PMID: 17082263 PMCID: PMC2932480 DOI: 10.1210/en.2006-0952] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
FSH is produced by the pituitary gonadotrope to regulate gametogenesis. Production of the beta-subunit of FSH is the rate-limiting step in FSH synthesis, and a number of peptide and steroid hormones within the reproductive axis have been found to regulate transcription of the FSH beta-subunit gene. Although both activin and glucocorticoids are notable regulators of FSHbeta by themselves, we find that cotreatment results in a synergistic interaction on the mouse FSHbeta promoter at the level of the gonadotrope using transient transfection of a reporter gene into the LbetaT2 immortalized gonadotrope-derived cell line. This synergistic interaction is specific to FSHbeta, because only additive effects of these two hormones are observed on LH beta-subunit, GnRH receptor, and mouse mammary tumor virus gene expression. Components of both activin and glucocorticoid signaling are found to be necessary for synergy, and there are specific cis elements on the mouse FSHbeta promoter that contribute to the synergistic response as well. We also identify novel activin-responsive regions in the mouse FSHbeta promoter and find that the -120 site can bind Smad2/3 in vitro. In addition, the glucocorticoid receptor and Smad3 are sufficient to confer a striking synergy with glucocorticoids on the mouse FSHbeta promoter. Our studies provide the first evidence of a synergistic interaction between activin and glucocorticoids within the gonadotrope cell and demonstrate that this synergy can occur directly at the level of the mouse FSHbeta promoter.
Collapse
Affiliation(s)
- Shauna M McGillivray
- Department of Reproductive Medicine, University of California at San Diego, 9500 Gilman Drive, La Jolla, California 92093-0674, USA
| | | | | | | |
Collapse
|
39
|
Mullen RD, Colvin SC, Hunter CS, Savage JJ, Walvoord EC, Bhangoo AP, Ten S, Weigel J, Pfäffle RW, Rhodes SJ. Roles of the LHX3 and LHX4 LIM-homeodomain factors in pituitary development. Mol Cell Endocrinol 2007; 265-266:190-5. [PMID: 17210222 PMCID: PMC1853274 DOI: 10.1016/j.mce.2006.12.019] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The LHX3 and LHX4 LIM-homeodomain transcription factors play essential roles in pituitary gland and nervous system development. Mutations in the genes encoding these regulatory proteins are associated with combined hormone deficiency diseases in humans and animal models. Patients with these diseases have complex syndromes involving short stature, and reproductive and metabolic disorders. Analyses of the features of these diseases and the biochemical properties of the LHX3 and LHX4 proteins will facilitate a better understanding of the molecular pathways that regulate the development of the specialized hormone-secreting cells of the mammalian anterior pituitary gland.
Collapse
Affiliation(s)
- Rachel D. Mullen
- Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
| | - Stephanie C. Colvin
- Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
- Biology, Indiana University-Purdue University, Indianapolis, IN
| | - Chad S. Hunter
- Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
- Biology, Indiana University-Purdue University, Indianapolis, IN
| | - Jesse J. Savage
- Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
- Biology, Indiana University-Purdue University, Indianapolis, IN
| | | | | | - Svetlana Ten
- Infants and Children’s Hospital of Brooklyn at Maimonides, Brooklyn, NY
| | | | | | - Simon J. Rhodes
- Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
40
|
Cherrington BD, Farmerie TA, Clay CM. A specific helical orientation underlies the functional contribution of the activin responsive unit to transcriptional activity of the murine gonadotropin-releasing hormone receptor gene promoter. Endocrine 2006; 29:425-33. [PMID: 16943581 DOI: 10.1385/endo:29:3:425] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2006] [Revised: 02/23/2006] [Accepted: 03/06/2006] [Indexed: 11/11/2022]
Abstract
Activin responsiveness of the murine GnRH receptor (GnRHR) gene promoter requires two spatially distinct regulatory elements termed the GnRH receptor activating sequence or GRAS and the downstream activin regulatory element or DARE. While GRAS interacts with multiple transcription factors, DARE activity requires tandem homeodomain binding motifs (TAAT) and displays specific binding to the LIM homeodomain protein LHX3. Herein, we find that both the murine GnRHR gene promoter and DARE fused to a minimal heterologous promoter are responsive to LHX3 overexpression. A dominant-repressor of LHX3 attenuates transcriptional activity of the murine GnRHR gene promoter but had no impact on activin responsiveness. Thus, LHX3 would not appear to be the protein mediating activin responsiveness of this promoter. Within DARE itself, the tandem TAAT motifs are separated by 4 bp. Although this arrangement differs from the prototypical P2 or P3 binding sites characterized for paired-like homeodomain proteins and from the directly abutting TAAT motifs found for LHX3, a LIM-class homeodomain protein, we find that separation of the TAAT sites by 5 and 10 bp decreases GnRHR promoter activity to a level similar to promoters containing loss of function mutations in either the proximal or distal TAAT motif. Thus, the juxtaposition of the TAAT sites is critical for the functional activity of DARE. That activin responsiveness of the GnRHR promoter requires both GRAS and DARE suggests that these elements may be both functionally and structurally coupled. As to the latter, GRAS and DARE are separated by 20 bp, thus placing the elements on the same side of the helical backbone. To determine if this spatial organization is functionally relevant, multiples of 5 bp were inserted or deleted between GRAS and DARE. Any insertion or deletion that resulted in a half-turn alteration in the helical positioning between the two elements reduced promoter activity. Thus, an important spatial relationship underlies functional cooperation between GRAS and DARE and the emergence of a complex activin responsive unit (ARU) within the mouse GnRHR promoter.
Collapse
Affiliation(s)
- Brian D Cherrington
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | | | | |
Collapse
|
41
|
Granger A, Bleux C, Kottler ML, Rhodes SJ, Counis R, Laverrière JN. The LIM-homeodomain proteins Isl-1 and Lhx3 act with steroidogenic factor 1 to enhance gonadotrope-specific activity of the gonadotropin-releasing hormone receptor gene promoter. Mol Endocrinol 2006; 20:2093-108. [PMID: 16613990 DOI: 10.1210/me.2005-0184] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The GnRH receptor (GnRH-R) plays a central role in mammalian reproductive function throughout adulthood. It also appears as an early marker gene of the presumptive gonadotrope lineage in developing pituitary. Here, using transient transfections combined with DNA/protein interaction assays, we have delineated cis-acting elements within the rat GnRH-R gene promoter that represent targets for the LIM-homeodomain (LIM-HD) proteins, Isl-1 and Lhx3. These factors, critical in early pituitary development, are thus also crucial for gonadotrope-specific expression of the GnRH-R gene. In heterologous cells, the expression of Isl-1 and Lhx3, together with steroidogenic factor 1 (SF-1), culminates in the activation of both the rat as well as human GnRH-R promoter, suggesting that this combination is evolutionarily conserved among mammals. The specificity of these LIM-HD factors is attested by the inefficiency of related proteins, including Lhx5 and Lhx9, to activate the GnRH-R gene promoter, as well as by the repressive capacity of a dominant-negative derivative of Lhx3. Accordingly, targeted deletion of the LIM response element decreases promoter activity. In addition, experiments with Gal4-SF-1 fusion proteins suggest that LIM-HD protein activity in gonadotrope cells is dependent upon SF-1 binding. Finally, using a transgenic model that allows monitoring of in vivo promoter activity, we show that the overlapping expression of Isl-1 and Lhx3 in the developing pituitary correlates with promoter activity. Collectively, these data suggest the occurrence of a specific LIM-HD pituitary code and designate the GnRH-R gene as the first identified transcriptional target of Isl-1 in the anterior pituitary.
Collapse
Affiliation(s)
- Anne Granger
- Physiologie de l'Axe Gonadotrope, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7079, Physiologie et Physiopathologie, Université Pierre et Marie Curie-Paris6, 4 place Jussieu, 75252 Paris cedex 05, France
| | | | | | | | | | | |
Collapse
|
42
|
Bhangoo APS, Hunter CS, Savage JJ, Anhalt H, Pavlakis S, Walvoord EC, Ten S, Rhodes SJ. Clinical case seminar: a novel LHX3 mutation presenting as combined pituitary hormonal deficiency. J Clin Endocrinol Metab 2006; 91:747-53. [PMID: 16394081 DOI: 10.1210/jc.2005-2360] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT LHX3 encodes LIM homeodomain class transcription factors with important roles in pituitary and nervous system development. The only previous report of LHX3 mutations described patients with two types of recessive mutations displaying combined pituitary hormone deficiency coupled with neck rigidity. OBJECTIVE We report a patient presenting a unique phenotype associated with a novel mutation in the LHX3 gene. PATIENT We report a 6-yr, 9-month-old boy born from a consanguineous relationship who presented shortly after birth with cyanosis, feeding difficulty, persistent jaundice, micropenis, and poor weight gain and growth rate. Laboratory data, including an undetectable TSH, low free T4, low IGF-I and IGF binding protein-3, prolactin deficiency, and LH and FSH deficiency were consistent with hypopituitarism. A rigid cervical spine leading to limited head rotation was noticed on follow-up examination. Magnetic resonance imaging revealed an apparently structurally normal cervical spine and a postcontrast hypointense lesion in the anterior pituitary. RESULTS Analysis of the LHX3 gene revealed homozygosity for a novel single-base-pair deletion in exon 2. This mutation leads to a frame shift predicted to result in the production of short, inactive LHX3 proteins. The results of in vitro translation experiments are consistent with this prediction. The parents of the patients are heterozygotes, indicating a recessive mode of action for the deletion allele. CONCLUSIONS The presence of a hypointense pituitary lesion and other clinical findings broadens the phenotype associated with LHX3 gene mutation.
Collapse
Affiliation(s)
- Amrit P S Bhangoo
- Pediatric Endocrinology Division, Infant's and Children's Hospital of Brooklyn at Maimonides, Brooklyn, New York 11219, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Yaden BC, Garcia M, Smith TPL, Rhodes SJ. Two promoters mediate transcription from the human LHX3 gene: involvement of nuclear factor I and specificity protein 1. Endocrinology 2006; 147:324-37. [PMID: 16179410 DOI: 10.1210/en.2005-0970] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The LHX3 transcription factor is required for pituitary and nervous system development in mammals. Mutations in the human gene are associated with hormone-deficiency diseases. The gene generates two mRNAs, hLHX3a and hLHX3b, which encode three proteins with different properties. Here, the cis elements and trans-acting factors that regulate the basal transcription of the two mRNAs are characterized. A comparative approach was taken featuring analysis of seven mammalian Lhx3 genes, with a focus on the human gene. Two conserved, TATA-less, GC-rich promoters that are used to transcribe the mRNAs precede exons 1a and 1b of hLHX3. Transcription start sites were mapped for both promoters. Deletion experiments showed most activity for reporter genes containing the basal promoters in the context of -2.0 kb of hLHX3a and 1.8 kb of intron 1a (hLHX3b). Transfection, site-directed mutation, electrophoretic mobility shift, Southwestern blot, and chromatin immunoprecipitation approaches were used to characterize the interaction of transcription factors with conserved elements in the promoters. Specificity protein 1 is a regulator of both promoters through interaction with GC boxes. In addition, a distal element within intron 1a that is recognized by nuclear factor I is critical for hLHX3b promoter function. We conclude that dual promoters allow regulated production of two hLHX3 mRNAs.
Collapse
Affiliation(s)
- Benjamin C Yaden
- Department of Biology (B.C.Y., M.G.), Indiana University-Purdue University Indianapolis, Indianapolis, Indiana 46202, USA
| | | | | | | |
Collapse
|
44
|
Hapgood JP, Sadie H, van Biljon W, Ronacher K. Regulation of expression of mammalian gonadotrophin-releasing hormone receptor genes. J Neuroendocrinol 2005; 17:619-38. [PMID: 16159375 DOI: 10.1111/j.1365-2826.2005.01353.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Gonadotrophin-releasing hormone (GnRH), acting via its cognate GnRH receptor (GnRHR), is the primary regulator of mammalian reproductive function, and hence GnRH analogues are extensively used in the treatment of hormone-dependent diseases, as well as for assisted reproductive techniques. In addition to its established endocrine role in gonadotrophin regulation in the pituitary, evidence is rapidly accumulating to support the expression and functional roles for two forms of GnRHR (GnRHR I and GnRHR II) in multiple and diverse extra-pituitary mammalian tissues and cells. These findings, together with findings indicating that mutations of the GnRHR are linked to the disease hypogonadotrophic hypogonadism and that GnRHRs play a direct role in neuronal migration and reproductive cancers, have presented new therapeutic targets and intensified research into the structure, function and mechanisms of regulation of expression of GnRHR genes. The present review focuses on the current knowledge on tissue-specific and hormonal regulation of transcription of mammalian GnRH receptor genes. Emerging insights, such as the discovery of diverse regulatory mechanisms in pituitary and extra-pituitary cell types, nonclassical mechanisms of steroid regulation, the use of composite elements for cell-specific expression, the increasing profile of hormones involved in regulation, the complexity of kinase pathways that target the GnRHR I gene, as well as species-differences, are highlighted. Although further research is necessary to understand the mechanisms of regulation of expression of GnRHR I and GnRHR II genes, the GnRHR is emerging as a potential target gene for facilitating cross-talk between neuroendocrine, immune and stress-response systems in multiple tissues via autocrine, paracrine and endocrine signalling.
Collapse
Affiliation(s)
- J P Hapgood
- Department of Biochemistry, University of Stellenbosch, Matieland, South Africa.
| | | | | | | |
Collapse
|