1
|
Wallis M, Xu Q, Krawczyk M, Skowronska-Krawczyk D. Evolution of the enhancer-rich regulatory region of the gene for the cell-type specific transcription factor POU1F1. Heliyon 2024; 10:e28640. [PMID: 38590853 PMCID: PMC10999999 DOI: 10.1016/j.heliyon.2024.e28640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 03/14/2024] [Accepted: 03/21/2024] [Indexed: 04/10/2024] Open
Abstract
Precise spatio-temporal expression of genes in organogenesis is regulated by the coordinated interplay of DNA elements such as promoter and enhancers present in the regulatory region of a given locus. POU1F1 transcription factor plays a crucial role in the development of somatotrophs, lactotrophs and thyrotrophs in the anterior pituitary gland, and in maintaining high expression of growth hormone, prolactin and TSH. In mouse, expression of POU1F1 is controlled by a region fenced by two CTCF sites, containing 5 upstream enhancer elements, designated E-A (5' to 3'). Elements C, B and A correspond to elements shown previously to play a role in pituitary development and hormonal expression; functional roles for elements E and D have not been reported. We performed comparative sequence analysis of this regulatory region and discovered that three elements, B, C and E, are present in all vertebrate groups except Agnatha. One very long (>2 kb) element (A) is unique to mammals suggesting a specific change in regulation of the gene in this group. Using DNA accessibility assay (ATAC-seq) we showed that conserved elements in anterior pituitary of four non-mammals are open, suggesting functionality as regulatory elements. We showed that, in many non-mammalian vertebrates, an additional upstream exon closely follows element E, leading to alternatively spliced transcripts. Here, element E functions as an alternative promoter, but in mammals this feature is lost, suggesting conversion of alternative promoter to enhancer. Our work shows that regulation of POU1F1 changed markedly during the course of vertebrate evolution, use of a low number of enhancer elements combined with alternative promoters in non-mammalian vertebrates being replaced by use of a unique combination of regulatory units in mammals. Most importantly, our work suggests that evolutionary conversion of alternate promoter to enhancer could be one of the evolutionary mechanisms of enhancer birth.
Collapse
Affiliation(s)
- Michael Wallis
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK
| | - Qianlan Xu
- Department of Physiology and Biophysics, Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, University of California, Irvine, CA, USA
| | - Michal Krawczyk
- Department of Physiology and Biophysics, Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, University of California, Irvine, CA, USA
| | - Dorota Skowronska-Krawczyk
- Department of Physiology and Biophysics, Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, University of California, Irvine, CA, USA
| |
Collapse
|
2
|
Nair SJ, Suter T, Wang S, Yang L, Yang F, Rosenfeld MG. Transcriptional enhancers at 40: evolution of a viral DNA element to nuclear architectural structures. Trends Genet 2022; 38:1019-1047. [PMID: 35811173 PMCID: PMC9474616 DOI: 10.1016/j.tig.2022.05.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/05/2022] [Accepted: 05/31/2022] [Indexed: 02/08/2023]
Abstract
Gene regulation by transcriptional enhancers is the dominant mechanism driving cell type- and signal-specific transcriptional diversity in metazoans. However, over four decades since the original discovery, how enhancers operate in the nuclear space remains largely enigmatic. Recent multidisciplinary efforts combining real-time imaging, genome sequencing, and biophysical strategies provide insightful but conflicting models of enhancer-mediated gene control. Here, we review the discovery and progress in enhancer biology, emphasizing the recent findings that acutely activated enhancers assemble regulatory machinery as mesoscale architectural structures with distinct physical properties. These findings help formulate novel models that explain several mysterious features of the assembly of transcriptional enhancers and the mechanisms of spatial control of gene expression.
Collapse
Affiliation(s)
- Sreejith J Nair
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20057, USA.
| | - Tom Suter
- Howard Hughes Medical Institute, Department and School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Susan Wang
- Howard Hughes Medical Institute, Department and School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Cellular and Molecular Medicine Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Lu Yang
- Howard Hughes Medical Institute, Department and School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Feng Yang
- Howard Hughes Medical Institute, Department and School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Michael G Rosenfeld
- Howard Hughes Medical Institute, Department and School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
3
|
Liu Y, Masternak MM, Schneider A, Zhi X. Dwarf mice as models for reproductive ageing research. Reprod Biomed Online 2021; 44:5-13. [PMID: 34794884 DOI: 10.1016/j.rbmo.2021.09.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/05/2021] [Accepted: 09/17/2021] [Indexed: 01/10/2023]
Abstract
Dwarf mice are characterized by extremely long lifespan, delayed ovarian ageing, altered metabolism, lower age-related oxidative damage and cancer incidence rate. Snell dwarf, Ames dwarf and growth hormone receptor knockout mice are three commonly used models. Despite studies focusing on ageing and metabolism, the reproductive features of female dwarf mice have also attracted interest over the last decade. Female Snell and Ames dwarf mice have regular oestrous cycles and ovulation rates, as in normal mice, but with a larger ovarian reserve and delayed ovarian ageing. The primordial follicle reserve in dwarf mice is greater than in normal littermates. Anti-Müllerian hormone (AMH) concentration is seven times higher in Ames dwarf mice than in their normal siblings, and ovarian transcriptomic profiling showed distinctive patterns in older Ames dwarf mice, especially enriched in inflammatory and immune response-related pathways. In addition, microRNA profiles also showed distinctive differences in Ames dwarf mice compared with normal control littermates. This review aims to summarize research progress on dwarf mice as models in the reproductive ageing field. Investigations focusing on the mechanisms of their reserved reproductive ability are much needed and are expected to provide additional molecular biological bases for the clinical practice of reproductive medicine in women.
Collapse
Affiliation(s)
- Yujun Liu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital Beijing, PR China; National Clinical Research Center for Obstetrics and Gynecology Beijing, PR China; Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education Beijing, PR China
| | - Michal M Masternak
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando FL, USA; Department of Head and Neck Surgery, Poznan University of Medical Sciences, Poznan, Poland
| | - Augusto Schneider
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Xu Zhi
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital Beijing, PR China; National Clinical Research Center for Obstetrics and Gynecology Beijing, PR China; Key Laboratory of Assisted Reproduction, Peking University, Ministry of Education Beijing, PR China.
| |
Collapse
|
4
|
Gergics P, Smith C, Bando H, Jorge AAL, Rockstroh-Lippold D, Vishnopolska SA, Castinetti F, Maksutova M, Carvalho LRS, Hoppmann J, Martínez Mayer J, Albarel F, Braslavsky D, Keselman A, Bergadá I, Martí MA, Saveanu A, Barlier A, Abou Jamra R, Guo MH, Dauber A, Nakaguma M, Mendonca BB, Jayakody SN, Ozel AB, Fang Q, Ma Q, Li JZ, Brue T, Pérez Millán MI, Arnhold IJP, Pfaeffle R, Kitzman JO, Camper SA. High-throughput splicing assays identify missense and silent splice-disruptive POU1F1 variants underlying pituitary hormone deficiency. Am J Hum Genet 2021; 108:1526-1539. [PMID: 34270938 DOI: 10.1016/j.ajhg.2021.06.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 06/18/2021] [Indexed: 12/13/2022] Open
Abstract
Pituitary hormone deficiency occurs in ∼1:4,000 live births. Approximately 3% of the cases are due to mutations in the alpha isoform of POU1F1, a pituitary-specific transcriptional activator. We found four separate heterozygous missense variants in unrelated individuals with hypopituitarism that were predicted to affect a minor isoform, POU1F1 beta, which can act as a transcriptional repressor. These variants retain repressor activity, but they shift splicing to favor the expression of the beta isoform, resulting in dominant-negative loss of function. Using a high-throughput splicing reporter assay, we tested 1,070 single-nucleotide variants in POU1F1. We identified 96 splice-disruptive variants, including 14 synonymous variants. In separate cohorts, we found two additional synonymous variants nominated by this screen that co-segregate with hypopituitarism. This study underlines the importance of evaluating the impact of variants on splicing and provides a catalog for interpretation of variants of unknown significance in POU1F1.
Collapse
Affiliation(s)
- Peter Gergics
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109-5618, USA
| | - Cathy Smith
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109-5618, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109-2218, USA
| | - Hironori Bando
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109-5618, USA
| | - Alexander A L Jorge
- Genetic Endocrinology Unit (LIM25), Division of Endocrinology, Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 01246-903, Brazil
| | - Denise Rockstroh-Lippold
- Department of Women's and Child Health, Division of Pediatric Endocrinology, University Hospital Leipzig, Leipzig 04103, Germany
| | - Sebastian A Vishnopolska
- Instituto de Biociencias, Biotecnología y Biología Traslacional, Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, CABA CE1428EHA, Argentina
| | - Frederic Castinetti
- Aix Marseille University, AP-HM, INSERM, Marseille Medical Genetics, Marmara Institute, La Conception Hospital, Department of Endocrinology, Marseille 13005, France
| | - Mariam Maksutova
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109-5618, USA
| | - Luciani Renata Silveira Carvalho
- Developmental Endocrinology Unit, Laboratory of Hormones and Molecular Genetics LIM/42, Division of Endocrinology, Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
| | - Julia Hoppmann
- Department of Women's and Child Health, Division of Pediatric Endocrinology, University Hospital Leipzig, Leipzig 04103, Germany
| | - Julián Martínez Mayer
- Instituto de Biociencias, Biotecnología y Biología Traslacional, Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, CABA CE1428EHA, Argentina
| | - Frédérique Albarel
- Aix Marseille University, AP-HM, INSERM, Marseille Medical Genetics, Marmara Institute, La Conception Hospital, Department of Endocrinology, Marseille 13005, France
| | - Debora Braslavsky
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá," FEI - CONICET - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Ciudad de Buenos Aires, CABA CE1428EHA, Argentina
| | - Ana Keselman
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá," FEI - CONICET - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Ciudad de Buenos Aires, CABA CE1428EHA, Argentina
| | - Ignacio Bergadá
- Centro de Investigaciones Endocrinológicas "Dr. César Bergadá," FEI - CONICET - División de Endocrinología, Hospital de Niños Ricardo Gutiérrez, Ciudad de Buenos Aires, CABA CE1428EHA, Argentina
| | - Marcelo A Martí
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires e Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales CONICET, Pabellòn 2 de Ciudad Universitaria, Ciudad de Buenos Aires, CABA C1428EHA, Argentina
| | - Alexandru Saveanu
- Aix Marseille University, AP-HM, INSERM, Marseille Medical Genetics, Marmara Institute, La Conception Hospital, Laboratory of Molecular Biology, Marseille 13385, France
| | - Anne Barlier
- Aix Marseille University, AP-HM, INSERM, Marseille Medical Genetics, Marmara Institute, La Conception Hospital, Laboratory of Molecular Biology, Marseille 13385, France
| | - Rami Abou Jamra
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig 04103, Germany
| | - Michael H Guo
- Division of Endocrinology, Boston Children's Hospital and Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Andrew Dauber
- Cincinnati Center for Growth Disorders, Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Marilena Nakaguma
- Developmental Endocrinology Unit, Laboratory of Hormones and Molecular Genetics LIM/42, Division of Endocrinology, Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
| | - Berenice B Mendonca
- Developmental Endocrinology Unit, Laboratory of Hormones and Molecular Genetics LIM/42, Division of Endocrinology, Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
| | - Sajini N Jayakody
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109-5618, USA
| | - A Bilge Ozel
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109-5618, USA
| | - Qing Fang
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109-5618, USA
| | - Qianyi Ma
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109-5618, USA
| | - Jun Z Li
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109-5618, USA
| | - Thierry Brue
- Aix Marseille University, AP-HM, INSERM, Marseille Medical Genetics, Marmara Institute, La Conception Hospital, Department of Endocrinology, Marseille 13005, France
| | - María Ines Pérez Millán
- Instituto de Biociencias, Biotecnología y Biología Traslacional, Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Ciudad de Buenos Aires, CABA CE1428EHA, Argentina
| | - Ivo J P Arnhold
- Developmental Endocrinology Unit, Laboratory of Hormones and Molecular Genetics LIM/42, Division of Endocrinology, Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-900, Brazil
| | - Roland Pfaeffle
- Department of Women's and Child Health, Division of Pediatric Endocrinology, University Hospital Leipzig, Leipzig 04103, Germany; Institute of Human Genetics, University of Leipzig Medical Center, Leipzig 04103, Germany
| | - Jacob O Kitzman
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109-5618, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109-2218, USA.
| | - Sally A Camper
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109-5618, USA.
| |
Collapse
|
5
|
Daly AZ, Dudley LA, Peel MT, Liebhaber SA, Parker SCJ, Camper SA. Multi-omic profiling of pituitary thyrotropic cells and progenitors. BMC Biol 2021; 19:76. [PMID: 33858413 PMCID: PMC8051135 DOI: 10.1186/s12915-021-01009-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 03/23/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The pituitary gland is a neuroendocrine organ containing diverse cell types specialized in secreting hormones that regulate physiology. Pituitary thyrotropes produce thyroid-stimulating hormone (TSH), a critical factor for growth and maintenance of metabolism. The transcription factors POU1F1 and GATA2 have been implicated in thyrotrope fate, but the transcriptomic and epigenomic landscapes of these neuroendocrine cells have not been characterized. The goal of this work was to discover transcriptional regulatory elements that drive thyrotrope fate. RESULTS We identified the transcription factors and epigenomic changes in chromatin that are associated with differentiation of POU1F1-expressing progenitors into thyrotropes using cell lines that represent an undifferentiated Pou1f1 lineage progenitor (GHF-T1) and a committed thyrotrope line that produces TSH (TαT1). We compared RNA-seq, ATAC-seq, histone modification (H3K27Ac, H3K4Me1, and H3K27Me3), and POU1F1 binding in these cell lines. POU1F1 binding sites are commonly associated with bZIP transcription factor consensus binding sites in GHF-T1 cells and Helix-Turn-Helix (HTH) or basic Helix-Loop-Helix (bHLH) factors in TαT1 cells, suggesting that these classes of transcription factors may recruit or cooperate with POU1F1 binding at unique sites. We validated enhancer function of novel elements we mapped near Cga, Pitx1, Gata2, and Tshb by transfection in TαT1 cells. Finally, we confirmed that an enhancer element near Tshb can drive expression in thyrotropes of transgenic mice, and we demonstrate that GATA2 enhances Tshb expression through this element. CONCLUSION These results extend the ENCODE multi-omic profiling approach to the pituitary gland, which should be valuable for understanding pituitary development and disease pathogenesis.
Collapse
Affiliation(s)
- Alexandre Z Daly
- Department Human Genetics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Lindsey A Dudley
- Department Human Genetics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Michael T Peel
- Department Genetics, University of Pennsylvania Perelman School of Medicine, Ann Arbor, MI, 48109, USA.,Incyte, Wilmington, DE, 19803, USA
| | - Stephen A Liebhaber
- Department Human Genetics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.,Department Genetics, University of Pennsylvania Perelman School of Medicine, Ann Arbor, MI, 48109, USA
| | - Stephen C J Parker
- Department Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Sally A Camper
- Department Human Genetics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
6
|
Ozaki H, Suga H, Arima H. Hypothalamic-pituitary organoid generation through the recapitulation of organogenesis. Dev Growth Differ 2021; 63:154-165. [PMID: 33662152 DOI: 10.1111/dgd.12719] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 11/26/2022]
Abstract
This paper overviews the development and differentiation of the hypothalamus and pituitary gland from embryonic stem (ES) and induced pluripotent stem (iPS) cells. It is important to replicate the developmental process in vivo to create specific cells/organoids from ES/iPS cells. We also introduce the latest findings and discuss future issues for clinical application. Neuroectodermal progenitors are induced from pluripotent stem cells by strictly removing exogenous patterning factors during the early differentiation period. The induced progenitors differentiate into rostral hypothalamic neurons, in particular magnocellular vasopressin+ neurons. In three-dimensional cultures, ES/iPS cells differentiate into hypothalamic neuroectoderm and nonneural head ectoderm adjacently. Rathke's pouch-like structures self-organize at the interface between the two layers and generate various endocrine cells, including corticotrophs and somatotrophs. Our next objective is to sophisticate our stepwise methodology to establish a novel transplantation treatment for hypopituitarism and apply it to developmental disease models.
Collapse
Affiliation(s)
- Hajime Ozaki
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Arima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
7
|
Hibara A, Yamaguchi T, Kojima M, Yamano Y, Higuchi M. Nicotine inhibits expression of Prrx1 in pituitary stem/progenitor cells through epigenetic regulation, leading to a delayed supply of growth-hormone-producing cells. Growth Horm IGF Res 2020; 51:65-74. [PMID: 32146343 DOI: 10.1016/j.ghir.2020.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 12/27/2019] [Accepted: 02/17/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Nicotine, a toxic component of smoking, adversely affects animal growth and reproduction by decreasing secretion of anterior pituitary hormones. However, it has not been clarified whether nicotine inhibits the supply of endocrine cells in the pituitary gland. The present study investigated short- and long-term effects of persistent nicotine exposure on the pituitary glands of young animals. DESIGN Three-week-old male Wistar rats were exposed to nicotine (1 mg/kg body weight/day) for 7 days, and gene expression, cell numbers, and DNA methylation status were analyzed on the following day and 4 weeks after final treatments. RESULTS The expression level of the stem cell marker Sox2 was not changed by nicotine exposure throughout the experiment. On the other hand, nicotine inhibited expression of a progenitor cell marker, Prrx1, and growth hormone (Gh). Immunohistochemical analysis showed that the SOX2-positive cells positive for PRRX1 in nicotine-treated groups decreased to 61% (4-week-old) and 70% (8-week-old) of the saline-treated controls. In addition, the proportion of GH-positive cells in nicotine-treated group was 14% lower than that of saline-treated controls. Furthermore, first intron hypermethylation of Prrx1 was detected by a bisulfite sequence of genomic DNA from the anterior lobe of the rat pituitary gland. CONCLUSIONS We show that persistent nicotine exposure in young animals inhibits expression of Prrx1 in pituitary stem/progenitor cells through epigenetic regulation, leading to a delayed supply of GH-producing cells.
Collapse
Affiliation(s)
- Ayaka Hibara
- Laboratory of Veterinary Biochemistry, Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, 4-101 Koyama-minami, Tottori-shi, Tottori 680-8553, Japan
| | - Takahiro Yamaguchi
- Laboratory of Veterinary Biochemistry, Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, 4-101 Koyama-minami, Tottori-shi, Tottori 680-8553, Japan
| | - Miki Kojima
- Laboratory of Veterinary Biochemistry, Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, 4-101 Koyama-minami, Tottori-shi, Tottori 680-8553, Japan
| | - Yoshiaki Yamano
- Laboratory of Veterinary Biochemistry, Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, 4-101 Koyama-minami, Tottori-shi, Tottori 680-8553, Japan
| | - Masashi Higuchi
- Laboratory of Veterinary Biochemistry, Joint Department of Veterinary Medicine, Faculty of Agriculture, Tottori University, 4-101 Koyama-minami, Tottori-shi, Tottori 680-8553, Japan.
| |
Collapse
|
8
|
Suga H. Application of pluripotent stem cells for treatment of human neuroendocrine disorders. Cell Tissue Res 2018; 375:267-278. [PMID: 30078102 DOI: 10.1007/s00441-018-2880-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 06/28/2018] [Indexed: 10/28/2022]
Abstract
The neuroendocrine system is composed of many types of functional cells. Matured cells are generally irreversible to progenitor cells and it is difficult to obtain enough from our body. Therefore, studying specific subtypes of human neuroendocrine cells in vitro has not been feasible. One of the solutions is pluripotent stem cells, such as embryonic stem (ES) cells and induced pluripotent stem (iPS) cells. These are unlimited sources and, in theory, are able to give rise to all cell types of our body. Therefore, we can use them for regenerative medicine, developmental basic research and disease modeling. Based on this idea, differentiation methods have been studied for years. Recent studies have successfully induced hypothalamic-like progenitors from mouse and human ES/iPS cells. The induced hypothalamic-like progenitors generated hypothalamic neurons, for instance, vasopressin neurons. Induction to adenohypophysis was also reported in the manner of self-formation by three-dimensional floating cultures. Rathke's pouch-like structures, i.e., pituitary anlage, were self-organized in accordance with pituitary development in embryo. Pituitary hormone-producing cells were subsequently differentiated. The induced corticotrophs secreted adrenocorticotropic hormone in response to corticotropin-releasing hormone. When engrafted in vivo, these cells rescued systemic glucocorticoid levels in hypopituitary mice. These culture methods were characterized by replication of stepwise embryonic differentiation. It is based on the idea of mimicking the molecular environment of embryogenesis. Thanks to these improvements, these days, we can generate hormone-secreting neuroendocrine cells from pluripotent stem cells. The next problems that need to be solved are improving differentiation efficiency even further and structuring networks.
Collapse
Affiliation(s)
- Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, 466-8550, Japan.
| |
Collapse
|
9
|
Shaikh SR, Shaver PR, Shewchuk BM. High Fat Diet Dysregulates Hypothalamic-Pituitary Axis Gene Expression Levels which are Differentially Rescued by EPA and DHA Ethyl Esters. Mol Nutr Food Res 2018; 62:e1800219. [PMID: 29738112 DOI: 10.1002/mnfr.201800219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 04/20/2018] [Indexed: 11/06/2022]
Abstract
SCOPE Dietary fat composition can modulate gene expression in peripheral tissues in obesity. Observations of the dysregulation of growth hormone (GH) in obesity indicate that these effects extend to the hypothalamic-pituitary (H-P) axis. The authors thus determine whether specific high fat (HF) diets influence the levels of Gh and other key gene transcripts in the H-P axis. METHODS AND RESULTS C57BL/6 mice are fed a lean control diet or a HF diet in the absence or presence of OA, EPA, or DHA ethyl esters. Comparative studies are conducted with menhaden fish oil. The HF diet lowered pituitary Gh mRNA and protein levels, and cell culture studies reveal that elevated insulin and glucose can reduce Gh transcripts. Supplementation of the HF diet with OA, EPA, DHA, or menhaden fish oil do not improve pituitary Gh levels. The HF diet also impaired the levels of additional genes in the pituitary and hypothalamus, which are selectively rescued with EPA or DHA ethyl esters. The effects of EPA and DHA are more robust relative to fish oil. CONCLUSION A HF diet can affect H-P axis transcription, which can be mitigated in some genes by EPA and DHA, but not fish oil in most cases.
Collapse
Affiliation(s)
- Saame Raza Shaikh
- Department of Biochemistry and Molecular Biology and the East Carolina University Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC, 27834.,Department of Nutrition, Gillings School of Global Public Health and School of Medicine, University of North Carolina at Chapel Hill, CB #7461, Chapel Hill, NC, 27599
| | - Patti R Shaver
- Department of Biochemistry and Molecular Biology and the East Carolina University Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC, 27834
| | - Brian M Shewchuk
- Department of Biochemistry and Molecular Biology and the East Carolina University Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, NC, 27834
| |
Collapse
|
10
|
Clump formation in mouse pituitary-derived non-endocrine cell line Tpit/F1 promotes differentiation into growth-hormone-producing cells. Cell Tissue Res 2017; 369:353-368. [DOI: 10.1007/s00441-017-2603-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 02/24/2017] [Indexed: 01/08/2023]
|
11
|
Abstract
The hypothalamic-pituitary system is essential for maintaining life and controlling systemic homeostasis. The functional disorder makes patients suffer from various symptoms all their lives. Pluripotent stem cells, such as embryonic stem (ES) cells and induced pluripotent stem (iPS) cells, differentiate into neuroectodermal progenitors when cultured as floating aggregates under serum-free conditions. Recent results have shown that strict removal of exogenous patterning factors during the early differentiation period induces rostral hypothalamic-like progenitors from mouse ES cells. The use of growth factor-free, chemically defined medium was critical for this induction. The ES cell-derived hypothalamic-like progenitors generated rostral-dorsal hypothalamic neurons, in particular magnocellular vasopressinergic neurons. We subsequently reported self-formation of adenohypophysis in three-dimensional floating cultures of mouse ES cells. The ES cell aggregates were stimulated to differentiate into both non-neural head ectoderm and hypothalamic neuroectoderm in adjacent layers. Self-organization of Rathke's pouch-like structures occurred at the interface of the two epithelia in vitro. Various pituitary endocrine cells including corticotrophs and somatotrophs were subsequently produced from the Rathke's pouch-like structures. The induced corticotrophs efficiently secreted ACTH in response to CRH. Furthermore, when engrafted in vivo, these cells rescued systemic glucocorticoid levels in hypopituitary mice. Our latest study aimed to prepare hypothalamic and pituitary tissues from human pluripotent stem cells. We succeeded in establishing the differentiation method using human ES/iPS cells. The culture method is characterized by replication of stepwise embryonic differentiation. Therefore, these methods could potentially be used as developmental and disease models, as well as for future regenerative medicine.
Collapse
Affiliation(s)
- Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Hospital, Nagoya 466-8550, Japan
| |
Collapse
|
12
|
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
|
13
|
An autoregulatory pathway establishes the definitive chromatin conformation at the pit-1 locus. Mol Cell Biol 2015; 35:1523-32. [PMID: 25691665 DOI: 10.1128/mcb.01283-14] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 02/09/2015] [Indexed: 12/12/2022] Open
Abstract
The transcription factor Pit-1 (POU1-F1) plays a dominant role in cell lineage expansion and differentiation in the anterior pituitary. Prior studies of the mouse Pit-1 (mPit-1) gene revealed that this master regulatory locus is activated at embryonic day 13.5 (E13.5) by an early enhancer (EE), whereas its subsequent expression throughout adult life is maintained by a more distal definitive enhancer (DE). Here, we demonstrate that the sequential actions of these two enhancers are linked to corresponding shifts in their proximities to the Pit-1 promoter. We further demonstrate that the looping of the definitive enhancer to the mPit-1 promoter is critically dependent on a self-sustaining autoregulatory mechanism mediated by the Pit-1 protein. These Pit-1-dependent actions are accompanied by localized recruitment of CBP and enrichment for H3K27 acetylation within the Pit-1 locus. These data support a model in which the sequential actions of two developmentally activated enhancers are linked to a corresponding shift in higher-order chromatin structures. This shift establishes an autoregulatory circuit that maintains durable expression of Pit-1 throughout adult life.
Collapse
|
14
|
Suga H. Differentiation of pluripotent stem cells into hypothalamic and pituitary cells. Neuroendocrinology 2015; 101:18-24. [PMID: 25428763 DOI: 10.1159/000369821] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 11/05/2014] [Indexed: 11/19/2022]
Abstract
The hypothalamic-pituitary system is essential to maintain life and control systemic homeostasis, but it is negatively affected by various diseases, leading to serious symptoms. Embryonic stem (ES) cells differentiate into neuroectodermal progenitors when cultured as floating aggregates under serum-free conditions. Recently, our colleagues have shown that strict removal of exogenous patterning factors during early differentiation steps induced efficient generation of rostral hypothalamic-like progenitors from mouse ES cell-derived neuroectodermal cells. The use of growth factor-free chemically defined medium was critical for this induction. The ES cell-derived hypothalamic-like progenitors generated rostral-dorsal hypothalamic neurons, especially magnocellular vasopressinergic neurons that release the hormone upon stimulation. Subsequently, we reported efficient self-formation of 3-dimensional adenohypophysis tissues in aggregate cultures of mouse ES cells. The ES cells were stimulated to differentiate into nonneural head ectoderm and hypothalamic neuroectoderm in adjacent layers within the aggregate and then treated with hedgehog. Self-organization of Rathke's pouch-like structures occurred at the interface of the two epithelia, as observed in vivo, and various endocrine cells including corticotrophs and somatotrophs were subsequently produced. The corticotrophs efficiently secreted adrenocorticotropic hormone in response to corticotropin-releasing hormone. Furthermore, when engrafted in vivo, these cells rescued the systemic glucocorticoid level in hypopituitary mice. Our present research aims are to prepare hypothalamic and pituitary tissues from human induced pluripotent stem cells and establish effective transplantation techniques with clinical applications. To replicate the complex and precise control of the hypothalamic-pituitary system, regenerative medicine using pluripotent cells may be a hopeful option.
Collapse
Affiliation(s)
- Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Hospital, Nagoya, Japan
| |
Collapse
|
15
|
Distinct chromatin configurations regulate the initiation and the maintenance of hGH gene expression. Mol Cell Biol 2013; 33:1723-34. [PMID: 23428872 DOI: 10.1128/mcb.01166-12] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
For many mammalian genes, initiation of transcription during embryonic development must be subsequently sustained over extensive periods of adult life. It remains unclear whether maintenance of gene expression reflects the same set of pathways as are involved in initial gene activation. The human pituitary growth hormone (hGH-N) locus is activated in the differentiating somatotrope midway through embryogenesis by a multicomponent locus control region (LCR). DNase I-hypersensitive site I (HSI) of the LCR is essential to full developmental activation of the hGH-N locus. Here we demonstrate that conditional deletion of HSI from the active hGH locus in the adult pituitary effectively silences hGH-N expression. Analyses of chromatin structure and locus positioning demonstrate that a specific subset of the HSI functions active in the embryo retain their HSI dependence in the adult pituitary. These functions sustain engagement of the hGH locus with polymerase II (Pol II) factories, histone acetylation at the hGH-N promoter, and looping of the LCR to its target promoter. These data reveal that HSI is essential to both the maintenance and the initiation phases of gene expression. These observations contribute to our mechanistic understanding of how stable patterns of mammalian gene expression are established in a terminally differentiated cell.
Collapse
|
16
|
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
|
17
|
POU1F1-mediated activation of hGH-N by deoxyribonuclease I hypersensitive site II of the human growth hormone locus control region. J Mol Biol 2011; 415:29-45. [PMID: 22094313 DOI: 10.1016/j.jmb.2011.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 10/21/2011] [Accepted: 11/01/2011] [Indexed: 11/23/2022]
Abstract
The human growth hormone gene (hGH-N) is regulated by a distal locus control region (LCR) composed of five deoxyribonuclease I hypersensitive sites (HSs). The region encompassing HSI and HSII contains the predominant pituitary somatotrope-specific hGH-N activation function of the LCR. This activity was attributed primarily to POU1F1 (Pit-1) elements at HSI, as linkage to HSI was sufficient for properly regulated hGH-N expression in transgenic mice, while HSII alone had no activity. However, the presence of HSII in conjunction with HSI further enhanced hGH-N transgene expression, indicating additional determinants of pituitary hGH-N activation in the HSII region, but limitations of transgenic models and previous ex vivo systems have prevented the characterization of HSII. In the present study, we employ a novel minichromosome model of the hGH-N regulatory domain and show that HSII confers robust POU1F1-dependent activation of hGH-N in this system. This effect was accompanied by POU1F1-dependent histone acetylation and methylation throughout the minichromosome LCR/hGH-N domain. A series of in vitro DNA binding experiments revealed that POU1F1 binds to multiple sites at HSII, consistent with a direct role in HSII function. Remarkably, POU1F1 binding was localized in part to the 3' untranslated region of a primate-specific LINE-1 (long interspersed nuclear element 1) retrotransposon, suggesting that its insertion during primate evolution may have conferred function to the HSII region in the context of pituitary GH gene regulation. These observations clarify the function of HSII, expanding the role of POU1F1 in hGH LCR activity, and provide insight on the molecular evolution of the LCR.
Collapse
|
18
|
Angotzi AR, Mungpakdee S, Stefansson S, Male R, Chourrout D. Involvement of Prop1 homeobox gene in the early development of fish pituitary gland. Gen Comp Endocrinol 2011; 171:332-40. [PMID: 21362424 DOI: 10.1016/j.ygcen.2011.02.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 02/17/2011] [Accepted: 02/21/2011] [Indexed: 11/22/2022]
Abstract
When mutated in mammals, paired-like homeobox Prop1 gene produces highly variable pituitary phenotypes with impaired regulation of Pit1 and eventually defective synthesis of Pit1-regulated pituitary hormones. Here we have identified fish prop1 orthologs, confirmed their pituitary-specific expression, and blocked the splicing of zebrafish prop1 transcripts using morpholino oligonucleotides. Very early steps of the gland formation seemed unaffected based on morphology and expression of early placodal marker pitx. Prop1 knock-down reduced the expression of pit1, prl (prolactin) and gh (growth hormone), as expected if the function of Prop1 is conserved throughout vertebrates. Less expectedly, lim3 was down regulated. This gene is expressed from early stages of vertebrate pituitary development but is not known to be Prop1-dependent. In situ hybridizations on prop1 morphants using probes for the pan pituitary gene pitx3 and for the hormone gene markers prl, gh and tshβ, revealed abnormal shape, growth and cellular organization of the developed adenohypophysis. Strikingly, the effects of prop1 knock-down on adenohypophysis morphology and gene expression were gradually reversed during late development, despite persistent splice-blocking of transcripts. Therefore, prop1 function appears to be conserved between mammals and fish, at least for the mediation of hormonal cell type differentiation via pit1, but the existence of other fish-specific pathways downstream of prop1 are suggested by our observations.
Collapse
Affiliation(s)
- Anna Rita Angotzi
- Sars, International Centre for Marine Molecular Biology, University of Bergen, High Technology Centre, Thormoehlensgt 55, N-5008 Bergen, Norway
| | | | | | | | | |
Collapse
|
19
|
Tajima T, Ohtake A, Hoshino M, Amemiya S, Sasaki N, Ishizu K, Fujieda K. OTX2 loss of function mutation causes anophthalmia and combined pituitary hormone deficiency with a small anterior and ectopic posterior pituitary. J Clin Endocrinol Metab 2009; 94:314-9. [PMID: 18854396 DOI: 10.1210/jc.2008-1219] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CONTEXT Orthodenticle homeobox 2 (OTX2) is a transcription factor necessary for ocular and forebrain development. In humans, heterozygous mutations of OTX2 cause severe ocular malformations. However, whether mutations of OTX2 cause pituitary structural abnormalities or combined pituitary hormone deficiency (CPHD) has not been clarified. OBJECTIVES We surveyed the functional consequences of a novel OTX2 mutation that was detected in a patient with anophthalmia and CPHD. PATIENT We examined a Japanese patient with growth disturbance, anophthalamia, and severe developmental delay. He showed deficiencies in GH, TSH, LH, FSH, and ACTH. Brain magnetic resonance imaging revealed a small anterior pituitary gland, invisible stalk, ectopic posterior lobe, and Chiari malformation. RESULTS Sequence analysis of OTX2 demonstrated a heterozygous two bases insertion [S136fsX178 (c.576-577insCT)] in exon 3. The mutant Otx2 protein localized to the nucleus, but did not activate the promoter of the HESX1 and POU1F1 gene, indicating a loss of function mutation. No dominant negative effect in the presence of wild-type Otx2 was observed. CONCLUSION This case indicates that the OTX2 mutation is a cause of CPHD. Further study of more patients with OTX2 defects is necessary to clarify the clinical phenotypes and endocrine defects caused by OTX2 mutations.
Collapse
Affiliation(s)
- Toshihiro Tajima
- Department of Pediatrics, Hokkaido University School of Medicine, N15, W7, Sapporo, Japan 060-0835.
| | | | | | | | | | | | | |
Collapse
|
20
|
Ikeshita N, Kawagishi M, Shibahara H, Toda K, Yamashita T, Yamamoto D, Sugiyama Y, Iguchi G, Iida K, Takahashi Y, Kaji H, Chihara K, Okimura Y. Identification and analysis of prophet of Pit-1-binding sites in human Pit-1 gene. Endocrinology 2008; 149:5491-9. [PMID: 18653712 DOI: 10.1210/en.2008-0030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Prophet of Pit-1 (Prop1) is a transcription factor that regulates Pit-1 gene expression. Because Pit-1 regulates the differentiation of pituitary cells and the expressions of GH, prolactin and TSHbeta genes, Prop1 mutation results in combined pituitary hormone deficiency in humans. However, Prop1-binding sites in human Pit-1 gene and the mechanism leading to combined pituitary hormone deficiency have remained unclear. In this study, we identified and analyzed Prop1-binding elements of the human Pit-1 gene. Prop1 stimulated the expression of the reporter plasmid containing Pit-1 gene from translation start site to -1340 dose dependently in GH3 cells. The activation by Prop1 was observed in GH3 and TtT/GF cells but not COS7, HeLa, JEG3, and HuH7 cells. Deletion analysis of Pit-1 gene showed that the Prop1-responsive elements were present within the -257-bp region. Within the -257-bp region, there are four elements similar to consensus sequence of paired-like transcription factors. Because Prop1 is a member of paired-like transcription factors, we assessed the elements. EMSA and transient transfection assay using the mutation of the elements revealed that the element from -63 to -53 (the proximal Prop1 binding element) was essential to Prop1-binding and Prop1-induced activation of Pit-1 reporter plasmid. A region at -8kb of human Pit-1 gene is similar to the distal region containing Prop1-binding elements in mouse Pit-1 gene. We showed the region functioned as an enhancer. Furthermore, chromatin immunoprecipitation assay showed that the proximal element could bind Prop1 in vivo cultured cells. Taken together, these findings indicated the novel functioning binding elements of Prop1 in human Pit-1 gene.
Collapse
Affiliation(s)
- Nobuko Ikeshita
- Department of Basic Allied Medicine, Kobe University School of Medicine, Kobe, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Egashira N, Minematsu T, Miyai S, Takekoshi S, Camper SA, Osamura RY. Pituitary changes in Prop1 transgenic mice: hormone producing tumors and signet-ring type gonadotropes. Acta Histochem Cytochem 2008; 41:47-57. [PMID: 18636109 PMCID: PMC2447862 DOI: 10.1267/ahc.08007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2008] [Accepted: 03/25/2008] [Indexed: 11/22/2022] Open
Abstract
Prophet of Pit-1 (Prop1) is an early transcription factor that delays the appearance of gonadotropin in the developing pituitaries. Prop1 transgenic (Tg) mice have been shown to generate pituitary tumors that either produce TSH or are non-hormone producing. In our series of Prop1 Tg mice, only 5 out of 9 female mice produced pituitary adenomas, and the adenomas were only GH, PRL, GH and PRL, PRL and gonadotropin or TSH producing. The pituitary cells that surrounded these adenomas showed hyperplasia of the corresponding hormone producing cells; i.e. the GH cells were increased in the pituitary that contained GH producing adenoma. In addition, although the adenomas lacked the expression of Prop1, the non-neoplastic pituitary cells showed expression of Prop1. The Prop1 Tg mice also showed vacuolated cells with eccentric nuclei, which are characteristic of “signet-ring hypertrophic cells”. Using immunohistochemistry, these signet ring hypertrophic cells were found to be positive for gonadotropin. Taken together, our results suggest a (1) tumorigenic effect of Prop1 in the pituitaries, and (2) causative effects of signet ring-type gonadotropes.
Collapse
Affiliation(s)
- Noboru Egashira
- Department of Pathology, Tokai University School of Medicine
| | - Takeo Minematsu
- Department of Pathology, Tokai University School of Medicine
| | - Syunsuke Miyai
- Department of Pathology, Tokai University School of Medicine
| | | | - Sally A. Camper
- Department of Human Genetics, University of Michigan Medical School
| | | |
Collapse
|
22
|
Qi Y, Ranish JA, Zhu X, Krones A, Zhang J, Aebersold R, Rose DW, Rosenfeld MG, Carrière C. Atbf1 is required for the Pit1 gene early activation. Proc Natl Acad Sci U S A 2008; 105:2481-6. [PMID: 18272476 PMCID: PMC2268162 DOI: 10.1073/pnas.0712196105] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Indexed: 12/24/2022] Open
Abstract
Enhancers have been functionally described for >35 years, but the molecular principles underlying the integration of regulatory inputs to alternate gene enhancers used during mammalian organogenesis remain incompletely understood. Using a combination of in vivo enhancer mapping and proteomics approaches, we have established that two distant and distinct early enhancers, each requiring different transcription complexes, are required for full activation of the gene encoding the pituitary lineage determining factor, Pit1. A transcription factor belonging to the "giant, multiple-homeodomain and zinc finger family," Atbf1, serves as a novel pituitary regulator for one of the two required enhancers as shown by genetic and in vitro analysis.
Collapse
Affiliation(s)
- Yingchuan Qi
- *Howard Hughes Medical Institute and Department of Medicine, School of Medicine, University of California at San Diego, La Jolla, CA 92093
| | - Jeffrey A. Ranish
- Institute for Systems Biology, 1441 North 34th Street, Seattle, WA 98103
| | - Xiaoyan Zhu
- *Howard Hughes Medical Institute and Department of Medicine, School of Medicine, University of California at San Diego, La Jolla, CA 92093
| | - Anna Krones
- *Howard Hughes Medical Institute and Department of Medicine, School of Medicine, University of California at San Diego, La Jolla, CA 92093
| | - Jie Zhang
- *Howard Hughes Medical Institute and Department of Medicine, School of Medicine, University of California at San Diego, La Jolla, CA 92093
| | - Ruedi Aebersold
- Institute for Systems Biology, 1441 North 34th Street, Seattle, WA 98103
- Institute of Molecular Systems Biology, Swiss Federal Institute of Technology (ETH) Hönggerberg and Faculty of Sciences, University of Zürich, CH-8093 Zürich, Switzerland
| | - David W. Rose
- Department of Medicine, Division of Endocrinology and Metabolism, School of Medicine, University of California at San Diego, La Jolla, CA 92093; and
| | - Michael G. Rosenfeld
- *Howard Hughes Medical Institute and Department of Medicine, School of Medicine, University of California at San Diego, La Jolla, CA 92093
| | - Catherine Carrière
- Department of Medicine, Dartmouth Medical School, Norris Cotton Cancer Center, 1 Medical Center Drive, Lebanon, NH 03756
| |
Collapse
|
23
|
Zhu X, Gleiberman AS, Rosenfeld MG. Molecular physiology of pituitary development: signaling and transcriptional networks. Physiol Rev 2007; 87:933-63. [PMID: 17615393 DOI: 10.1152/physrev.00006.2006] [Citation(s) in RCA: 238] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The pituitary gland is a central endocrine organ regulating basic physiological functions, including growth, the stress response, reproduction, metabolic homeostasis, and lactation. Distinct hormone-producing cell types in the anterior pituitary arise from a common ectodermal primordium during development by extrinsic and intrinsic mechanisms, providing a powerful model system for elucidating general principles in mammalian organogenesis. The central purpose of this review is to inspect the integrated signaling and transcriptional events that affect precursor proliferation, cell lineage commitment, terminal differentiation, and physiological regulation by hypothalamic tropic factors.
Collapse
Affiliation(s)
- Xiaoyan Zhu
- Howard Hughes Medical Institute, Department and School of Medicine, University of California, San Diego, La Jolla, California 92093, USA.
| | | | | |
Collapse
|
24
|
Olson LE, Tollkuhn J, Scafoglio C, Krones A, Zhang J, Ohgi KA, Wu W, Taketo MM, Kemler R, Grosschedl R, Rose D, Li X, Rosenfeld MG. Homeodomain-mediated beta-catenin-dependent switching events dictate cell-lineage determination. Cell 2006; 125:593-605. [PMID: 16678101 DOI: 10.1016/j.cell.2006.02.046] [Citation(s) in RCA: 210] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2005] [Revised: 12/13/2005] [Accepted: 02/09/2006] [Indexed: 11/24/2022]
Abstract
While the biological roles of canonical Wnt/beta-catenin signaling in development and disease are well documented, understanding the molecular logic underlying the functionally distinct nuclear transcriptional programs mediating the diverse functions of beta-catenin remains a major challenge. Here, we report an unexpected strategy for beta-catenin-dependent regulation of cell-lineage determination based on interactions between beta-catenin and a specific homeodomain factor, Prop1, rather than Lef/Tcfs. beta-catenin acts as a binary switch to simultaneously activate expression of the critical lineage-determining transcription factor, Pit1, and to repress the gene encoding the lineage-inhibiting transcription factor, Hesx1, acting via TLE/Reptin/HDAC1 corepressor complexes. The strategy of functionally distinct actions of a homeodomain factor in response to Wnt signaling is suggested to be prototypic of a widely used mechanism for generating diverse cell types from pluripotent precursor cells in response to common signaling pathways during organogenesis.
Collapse
Affiliation(s)
- Lorin E Olson
- Howard Hughes Medical Institute, Department and School of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Scott MM, Krueger KC, Deneris ES. A differentially autoregulated Pet-1 enhancer region is a critical target of the transcriptional cascade that governs serotonin neuron development. J Neurosci 2006; 25:2628-36. [PMID: 15758173 PMCID: PMC6725185 DOI: 10.1523/jneurosci.4979-04.2005] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The Pet-1 [pheochromocytoma 12 ETS (E26 transformation-specific)] gene plays a critical role in the development of serotonin (5-HT)-modulated behaviors via its control of embryonic 5-HT neuron differentiation. Pet-1 transcription is induced exclusively in 5-HT neuron postmitotic precursors before the appearance of transmitter, and its restricted expression is maintained in the adult. However, the mechanisms that direct Pet-1 expression to this single CNS neuronal cell type are unknown. Here, we show, using transgenic methods, that genomic sequences upstream, but not downstream or within the Pet-1-coding region, are sufficient for 5-HT neuron-specific transgene expression. Enhancer sequences within a 40 kb upstream fragment directed position-independent lacZ (beta-D-galactosidase) transgene expression to the developing hindbrain before the appearance of 5-HT. Moreover, virtually all of the 5-HT neurons in the adult were lacZ positive in all of the lines examined. Transgene expression in 5-HT neurons was maintained when the 40 kb fragment was truncated on its 5' end to either 12 or 1.8 kb, although position independence was then lost. Analysis of transgene expression in Pet-1 null mice indicated that Pet-1 was required to maintain the activity of the Pet-1 enhancer region in a subset of 5-HT neurons. These findings suggest that a conserved 1.8 kb region immediately flanking the Pet-1-coding region is a critical genomic target of the transcriptional cascade that governs 5-HT neuron development and provide additional evidence for 5-HT neuron heterogeneity at the genetic level. We discuss the potential application of the Pet-1 transgenes reported here to the selective genetic manipulation of 5-HT neurons.
Collapse
Affiliation(s)
- Michael M Scott
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | |
Collapse
|
26
|
Zhu X, Lin CR, Prefontaine GG, Tollkuhn J, Rosenfeld MG. Genetic control of pituitary development and hypopituitarism. Curr Opin Genet Dev 2005; 15:332-40. [PMID: 15917210 DOI: 10.1016/j.gde.2005.04.011] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2005] [Accepted: 04/12/2005] [Indexed: 10/25/2022]
Abstract
The pituitary gland functions as a relay between the hypothalamus and peripheral target organs that regulate basic physiological functions, including growth, the stress response, reproduction, metabolism and lactation. The development of the pituitary gland has been studied extensively in mice, and has begun to be explored in zebrafish, an animal model system amenable to forward genetics. Multiple signaling molecules and transcription factors, expressed in overlapping but distinct spatial and temporal patterns, are required at various stages of pituitary development. Defects in this precisely regulated genetic program lead to diverse pituitary dysfunction. The animal models have greatly enhanced our understanding of molecular mechanisms underlying pituitary development in addition to congenital pituitary disorders in humans.
Collapse
Affiliation(s)
- Xiaoyan Zhu
- Howard Hughes Medical Institute, Department and School of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | | | | | | | | |
Collapse
|
27
|
Suzuki K, Drevet J, Hinton BT, Huhtaniemi I, Lareyre JJ, Matusik RJ, Pons E, Poutanen M, Sipilä P, Orgebin-Crist MC. Epididymis-specific promoter-driven gene targeting: a new approach to control epididymal function? Mol Cell Endocrinol 2004; 216:15-22. [PMID: 15109740 DOI: 10.1016/j.mce.2003.10.070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Kichiya Suzuki
- Department of Obstetric and Gynecology, Vanderbilt Univerity School of Medicine, Nashville, TN 37232, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Savage JJ, Yaden BC, Kiratipranon P, Rhodes SJ. Transcriptional control during mammalian anterior pituitary development. Gene 2004; 319:1-19. [PMID: 14597167 DOI: 10.1016/s0378-1119(03)00804-7] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The mammalian anterior pituitary gland is a compound endocrine organ that regulates reproductive development and fitness, growth, metabolic homeostasis, the response to stress, and lactation, by actions on target organs such as the gonads, the liver, the thyroid, the adrenals, and the mammary gland. The protein and peptide hormones that control these physiological parameters are secreted by specialized pituitary cell types that derive from a common origin in the early ectoderm. Collectively, the broad physiological importance of the pituitary gland, its intriguing organogenesis, and the clinical and agricultural significance of its actions, have established pituitary development as an excellent model system for the study of the gene-regulatory cascades that guide vertebrate cell determination and differentiation. We review the transcriptional pathways that regulate the commitment of the individual pituitary cell lineages and that subsequently modulate trophic hormone gene activity in the differentiated cells of the mature gland.
Collapse
Affiliation(s)
- Jesse J Savage
- Department of Biology, Indiana University Center for Regenerative Biology and Medicine, Indiana University-Purdue University Indianapolis, 723 West Michigan Street, Indianapolis, IN 46202-5132, USA
| | | | | | | |
Collapse
|
29
|
Abstract
Whole-genome sequence assemblies are now available for seven different animals, including nematode worms, mice and humans. Comparative genome analyses reveal a surprising constancy in genetic content: vertebrate genomes have only about twice the number of genes that invertebrate genomes have, and the increase is primarily due to the duplication of existing genes rather than the invention of new ones. How, then, has evolutionary diversity arisen? Emerging evidence suggests that organismal complexity arises from progressively more elaborate regulation of gene expression.
Collapse
Affiliation(s)
- Michael Levine
- Department of Molecular and Cell Biology, Division of Genetics and Development, Center for Integrative Genomics, University of California, Berkeley, 401 Barker Hall, Berkeley, California 94720, USA.
| | | |
Collapse
|
30
|
Abstract
There is evidence that many peptide growth factors and hormones act in the intracellular space after either internalization or retention in their cells of synthesis. These factors, commonly called intracrines, are structurally diverse while sharing some common functional features. Reports of intracellular peptide hormone binding and action are reviewed here. Also, this laboratory has made proposals regarding the origin and actions of intracrines and these areas are further explored. Intracrine interactions and the relationship of intracrines to transcription factors are discussed. The intracellular/intracrine renin-angiotensin system (iRAS) is reviewed to illustrate the intracrine analogue of a well-established physiological system. The role of intracrine action in metazoan development is also considered.
Collapse
Affiliation(s)
- Richard N Re
- Research Division, Ochsner Clinic Foundation, 99 1514 Jefferson Highway, New Orleans, LA 70121, USA.
| |
Collapse
|
31
|
Trieu M, Ma A, Eng SR, Fedtsova N, Turner EE. Direct autoregulation and gene dosage compensation by POU-domain transcription factor Brn3a. Development 2003; 130:111-21. [PMID: 12441296 DOI: 10.1242/dev.00194] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Brn3a is a POU-domain transcription factor expressed in peripheral sensory neurons and in specific interneurons of the caudal CNS. Sensory expression of Brn3a is regulated by a specific upstream enhancer, the activity of which is greatly increased in Brn3a knockout mice, implying that Brn3a negatively regulates its own expression. Brn3a binds to highly conserved sites within this enhancer, and alteration of these sites abolishes Brn3a regulation of reporter transgenes. Furthermore, endogenous Brn3a expression levels in the sensory ganglia of Brn3a(+/+) and Brn3a(+/-) mice are similar, demonstrating that autoregulation can compensate for the loss of one allele by increasing transcription of the remaining gene copy. Conversely, transgenic overexpression of Brn3a in the trigeminal ganglion suppresses the expression of the endogenous gene. These findings demonstrate that the Brn3a locus functions as a self-regulating unit to maintain a constant expression level of this key regulator of neural development.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Base Sequence
- Central Nervous System/cytology
- Central Nervous System/embryology
- Central Nervous System/metabolism
- Cloning, Molecular
- Conserved Sequence
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Enhancer Elements, Genetic
- Ganglia, Sensory/cytology
- Ganglia, Sensory/embryology
- Ganglia, Sensory/metabolism
- Gene Dosage
- Gene Expression Regulation, Developmental
- Homeostasis
- Humans
- Mice
- Mice, Knockout
- Mice, Transgenic
- Molecular Sequence Data
- Neurons, Afferent/physiology
- Protein Structure, Tertiary
- Transcription Factor Brn-3
- Transcription Factor Brn-3A
- Transcription Factors/genetics
- Transcription Factors/metabolism
Collapse
Affiliation(s)
- May Trieu
- Department of Psychiatry, University of California, San Diego and San Diego VA Medical Center, La Jolla, CA 92093-0603, USA
| | | | | | | | | |
Collapse
|
32
|
Seuntjens E, Hauspie A, Vankelecom H, Denef C. Ontogeny of plurihormonal cells in the anterior pituitary of the mouse, as studied by means of hormone mRNA detection in single cells. J Neuroendocrinol 2002; 14:611-9. [PMID: 12153463 DOI: 10.1046/j.1365-2826.2002.00808.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The expression of mRNA of growth hormone (GH), prolactin (PRL), pro-opiomelanocortin (POMC) and the common glycoprotein hormone alpha-subunit (alphaGSU) was studied by means of single cell reverse transcriptase-polymerase chain reaction in male mouse pituitary cells at key time points of fetal and postnatal development: embryonic day 16 (E16); postnatal day 1 (P1) and young-adult age (P38). At E16, the hormone mRNAs examined were detectable, although only in 44% of total cells. Most of the hormone-positive cells expressed only one of the tested hormone mRNAs (monohormonal) but 14% of them contained more than one hormone mRNA (plurihormonal cells). Combinations of GH mRNA with PRL mRNA, of alphaGSU mRNA with GH and/or PRL mRNA and of POMC mRNA with GH and/or PRL mRNA or alphaGSU mRNA were found. As expected, the proportion of hormone-positive cells rose as the mouse aged. The proportions of plurihormonal cells followed a developmental pattern independent of that of monohormonal cells and characteristic for each hormone mRNA examined. Cells coexpressing POMC mRNA with GH or PRL mRNA significantly rose in proportion between E16 and P1, while the proportion of cells coexpressing GH and PRL mRNA markedly increased between P1 and P38. The occurrence of cells displaying combined expression of alphaGSU mRNA with GH and/or PRL mRNA did not significantly change during development. Remarkably, the population of cells expressing PRL mRNA only, was larger at E16 than at P1 and expanded again thereafter. In conclusion, the normal mouse pituitary develops a cell population that is capable of expressing multiple hormone mRNAs, thereby combining typical phenotypes of different cell lineages. These plurihormonal cells are already present during embryonic life. This population is of potential physiological relevance because development-related factors appear to determine which hormone mRNAs are preferentially coexpressed. Coexpression of multiple hormone mRNAs may represent a mechanism to respond to temporally increased endocrine demands. The data also suggest that the control of combined hormone expression is different from that of single hormone expression, raising questions about the current view on pituitary cell lineage specifications.
Collapse
Affiliation(s)
- E Seuntjens
- Laboratory of Cell Pharmacology, University of Leuven (KU Leuven), School of Medicine, Leuven, Belgium
| | | | | | | |
Collapse
|
33
|
Gerrish K, Cissell MA, Stein R. The role of hepatic nuclear factor 1 alpha and PDX-1 in transcriptional regulation of the pdx-1 gene. J Biol Chem 2001; 276:47775-84. [PMID: 11590182 DOI: 10.1074/jbc.m109244200] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The PDX-1 homeodomain transcription factor regulates pancreatic development and adult islet beta cell function. Expression of the pdx-1 gene is almost exclusively localized to beta cells within the adult endocrine pancreas. Islet beta cell-selective transcription is controlled by evolutionarily conserved subdomain sequences (termed Areas I (-2839 to -2520 base pairs (bp)), II (-2252 to -2023 bp), and III (-1939 to -1664 bp)) found within the 5'-flanking region of the pdx-1 gene. Areas I and II are independently capable of directing beta cell-selective reporter gene activity in transfection assays, with Area I-mediated stimulation dependent upon binding of hepatic nuclear factor 3 beta (HNF3 beta), a key regulator of islet beta cell function. To identify other transactivators of Area I, highly conserved sequence segments within this subdomain were mutagenized, and their effect on activation was determined. Several of the sensitive sites were found by transcription factor data base analysis to potentially bind endodermally expressed transcription factors, including HNF1 alpha (-2758 to -2746 bp, Segment 2), HNF4 (-2742 to -2730 bp, Segment 4; -2683 to -2671 bp, Segment 7-8), and HNF6 (-2727 to -2715 bp, Segment 5). HNF1 alpha, but not HNF4 and HNF6, binds specifically to Area I sequences in vitro. HNF1 alpha was also shown to specifically activate Area I-driven transcription through Segment 2. In addition, PDX-1 itself was found to stimulate Area I activation. The chromatin immunoprecipitation assay performed with PDX-1 antisera also demonstrated that this factor bound to Area I within the endogenous pdx-1 gene in beta cells. Our results indicate that regulatory factors binding to Area I conserved sequences contribute to the selective transcription pattern of the pdx-1 gene and that control is mediated by endodermal regulators like HNF1 alpha, HNF3 beta, and PDX-1.
Collapse
Affiliation(s)
- K Gerrish
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, Tennessee 37215, USA
| | | | | |
Collapse
|
34
|
Abstract
During the development of the pituitary gland, distinct hormone-producing cell types arise from a common population of ectodermal progenitors, providing an instructive model system for elucidating the molecular mechanisms of patterning and cell type specification in mammalian organogenesis. Recent studies have established that the development of the pituitary occurs through multiple sequential steps, allowing the coordinate control of the commitment, early patterning, proliferation, and positional determination of pituitary cell lineages in response to extrinsic and intrinsic signals. The early phases of pituitary development appear to be mediated through the activities of multiple signaling gradients emanating from key organizing centers that give rise to temporally and spatially distinct patterns of transcription factor expression. The induction of these transcriptional mediators in turn acts to positionally organize specific pituitary cell lineages within an apparently uniform field of ectodermal progenitors. Ultimately, pituitary cell types have proven to be both specified and maintained through the combinatorial interactions of a series of cell-type-restricted transcription factors that dictate the cell autonomous programs of differentiation in response to the transient signaling events.
Collapse
Affiliation(s)
- J S Dasen
- Howard Hughes Medical Institute, Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California 92093-0648, USA.
| | | |
Collapse
|
35
|
García A, Alvarez CV, Smith RG, Diéguez C. Regulation of Pit-1 expression by ghrelin and GHRP-6 through the GH secretagogue receptor. Mol Endocrinol 2001; 15:1484-95. [PMID: 11518797 DOI: 10.1210/mend.15.9.0694] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
GH secretagogues are an expanding class of synthetic peptide and nonpeptide molecules that stimulate the pituitary gland to secrete GH through their own specific receptor, the GH-secretagogue receptor. The cloning of the receptor for these nonclassical GH releasing molecules, together with the more recent characterization of an endogenous ligand, named ghrelin, have unambiguously demonstrated the existence of a physiological system that regulates GH secretion. Somatotroph cell-specific expression of the GH gene is dependent on a pituitary-specific transcription factor (Pit-1). This factor is transcribed in a highly restricted manner in the anterior pituitary gland. The present experiments sought to determine whether the synthetic hexapeptide GHRP-6, a reference GH secretagogue compound, as well as an endogenous ligand, ghrelin, regulate pit-1 expression. By a combination of Northern and Western blot analysis we found that GHRP-6 elicits a time- and dose-dependent activation of pit-1 expression in monolayer cultures of infant rat anterior pituitary cells. This effect was blocked by pretreatment with actinomycin D, but not by cycloheximide, suggesting that this action was due to direct transcriptional activation of pit-1. Using an established cell line (HEK293-GHS-R) that overexpresses the GH secretagogue receptor, we showed a marked stimulatory effect of GHRP-6 on the pit-1 -2,500 bp 5'-region driving luciferase expression. We truncated the responsive region to -231 bp, a sequence that contains two CREs, and found that both CREs are needed for GHRP-6-induced transcriptional activation in both HEK293-GHS-R cells and infant rat anterior pituitary primary cultures. The effect was dependent on PKC, MAPK kinase, and PKA activation. Increasing Pit-1 by coexpression of pCMV-pit-1 potentiated the GHRP-6 effect on the pit-1 promoter. Similarly, we showed that the endogenous GH secretagogue receptor ligand ghrelin exerts a similar effect on the pit-1 promoter. These data provide the first evidence that ghrelin, in addition to its previously reported GH-releasing activities, is also capable of regulating pit-1 transcription through the GH secretagogue receptor in the pituitary, thus giving new insights into the physiological role of the GH secretagogue receptor on somatotroph cell differentiation and function.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Cells, Cultured
- Cycloheximide/pharmacology
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Dactinomycin/pharmacology
- Dose-Response Relationship, Drug
- Gene Expression Regulation
- Genes, Reporter
- Ghrelin
- Growth Hormone/metabolism
- Humans
- Immunoblotting
- Ligands
- MAP Kinase Kinase 1
- Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors
- Mitogen-Activated Protein Kinase Kinases/metabolism
- Nucleic Acid Synthesis Inhibitors/pharmacology
- Oligopeptides/pharmacology
- Peptide Hormones
- Peptides/pharmacology
- Pituitary Gland, Anterior/cytology
- Pituitary Gland, Anterior/drug effects
- Pituitary Gland, Anterior/metabolism
- Prolactin/metabolism
- Protein Kinase C/antagonists & inhibitors
- Protein Kinase C/metabolism
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/metabolism
- Protein Synthesis Inhibitors/pharmacology
- Rats
- Receptors, Cell Surface/metabolism
- Receptors, G-Protein-Coupled
- Receptors, Ghrelin
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Regulatory Sequences, Nucleic Acid/genetics
- Transcription Factor Pit-1
- Transcription Factors/genetics
- Transcription Factors/metabolism
Collapse
Affiliation(s)
- A García
- Department of Physiology, Faculty of Medicine, University of Santiago de Compostela, Spain
| | | | | | | |
Collapse
|
36
|
Weatherly KL, Ramesh R, Strange H, Waite KL, Storrie B, Proudman JA, Wong EA. The turkey transcription factor Pit-1/GHF-1 can activate the turkey prolactin and growth hormone gene promoters in vitro but is not detectable in lactotrophs in vivo. Gen Comp Endocrinol 2001; 123:244-53. [PMID: 11589626 DOI: 10.1006/gcen.2001.7680] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The transcription factor Pit-1/GHF-1 plays an important role in regulating the prolactin (Prl) and growth hormone (GH) genes in mammals. In this study, the role that Pit-1 plays in regulating the prolactin and growth hormone genes in avian species was examined by cotransfection assays and immunofluorescence staining of pituitary sections. In cotransfection assays, turkey Pit-1 activated the turkey Prl, turkey GH, and rat Prl promoters 3.8-, 3.7-, and 12.5-fold, respectively. This activation was comparable to rat Pit-1 activation of these same promoters. A point mutation in the turkey Pit-1 cDNA, which changed leu-219 to ser-219, resulted in a 2-, 2-, and 10-fold reduction in the activation of the turkey Prl, turkey GH, and rat Prl promoters, respectively. Unexpectedly, coexpression of tPit-1 (leu-219) and tPit-1(ser-219) activated turkey Prl and rat Prl promoters 9.4- and 35.9-fold, respectively, but had no effect on the turkey GH promoter. Dual-label immunofluorescence analysis of turkey pituitary sections revealed that Pit-1 was not detectable in prolactin-staining cells but was detectable in GH-staining cells. Taken together, these data indicate that in the domestic turkey, Pit-1 can activate the turkey Prl promoter in vitro, but does not appear to play a role in regulating Prl gene expression in vivo. Pit-1, however, still likely plays a role in regulating GH gene expression.
Collapse
Affiliation(s)
- K L Weatherly
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, Virginia 24061-0306, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Thomas MK, Devon ON, Lee JH, Peter A, Schlosser DA, Tenser MS, Habener JF. Development of diabetes mellitus in aging transgenic mice following suppression of pancreatic homeoprotein IDX-1. J Clin Invest 2001. [DOI: 10.1172/jci200112029] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
38
|
Thomas MK, Devon ON, Lee JH, Peter A, Schlosser DA, Tenser MS, Habener JF. Development of diabetes mellitus in aging transgenic mice following suppression of pancreatic homeoprotein IDX-1. J Clin Invest 2001; 108:319-29. [PMID: 11457885 PMCID: PMC203024 DOI: 10.1172/jci12029] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Monogenic forms of diabetes can result from mutations in genes encoding transcription factors. Mutations in the homeodomain transcription factor IDX-1, a critical regulator of pancreas development and insulin gene transcription, confer a strong predisposition to the development of diabetes mellitus in humans. To investigate the role of IDX-1 expression in the pathogenesis of diabetes, we developed a model for the inducible impairment of IDX-1 expression in pancreatic beta cells in vivo by engineering an antisense ribozyme specific for mouse IDX-1 mRNA under control of the reverse tetracycline transactivator (rtTA). Doxycycline-induced impairment of IDX-1 expression reduced activation of the Insulin promoter but activated the Idx-1 promoter, suggesting that pancreatic beta cells regulate IDX-1 transcription to maintain IDX-1 levels within a narrow range. In transgenic mice that express both rtTA and the antisense ribozyme construct, impaired IDX-1 expression elevated glycated hemoglobin levels, diminished glucose tolerance, and decreased insulin/glucose ratios. Metabolic phenotypes induced by IDX-1 deficiency were observed predominantly in male mice over 18 months of age, suggesting that cellular mechanisms to protect IDX-1 levels in pancreatic beta cells decline with aging. We propose that even in the absence of Idx-1 gene mutations, pathophysiological processes that decrease IDX-1 levels are likely to impair glucose tolerance. Therapeutic strategies to attain normal glucose homeostasis by restoring normal IDX-1 levels may be of particular importance for older individuals with diabetes mellitus.
Collapse
Affiliation(s)
- M K Thomas
- Laboratory of Molecular Endocrinology, Massachusetts General Hospital, Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Kobayashi M, Nishikawa K, Yamamoto M. Hematopoietic regulatory domain of gata1 gene is positively regulated by GATA1 protein in zebrafish embryos. Development 2001; 128:2341-50. [PMID: 11493553 DOI: 10.1242/dev.128.12.2341] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Expression of gata1 is regulated through multiple cis-acting GATA motifs. To elucidate regulatory mechanisms of the gata1 gene, we have used zebrafish. To this end, we isolated and analyzed zebrafish gata1 genomic DNA, which resulted in the discovery of a novel intron that was unknown in previous analyses. This intron corresponds to the first intron of other vertebrate Gata1 genes. GFP reporter analyses revealed that this intron and a distal double GATA motif in the regulatory region are important for the regulation of zebrafish gata1 gene expression. To examine whether GATA1 regulates its own gene expression, we microinjected into embryos a GFP reporter gene linked successively to the gata1 gene regulatory region and to GATA1 mRNA. Surprisingly, ectopic expression of the reporter gene was induced at the site of GATA1 overexpression and was dependent on the distal double GATA motif. Functional domain analyses using transgenic fish lines that harbor the gata1-GFP reporter construct revealed that both the N- and C-terminal zinc-finger domains of GATA1, hence intact GATA1 function, are required for the ectopic GFP expression. These results provide the first in vivo evidence that gata1 gene expression undergoes positive autoregulation.
Collapse
Affiliation(s)
- M Kobayashi
- Center for Tsukuba Advanced Research Alliance and Institute of Basic Medical Sciences, University of Tsukuba, Tsukuba 305-8577, Japan
| | | | | |
Collapse
|
40
|
Andersen B, Rosenfeld MG. POU domain factors in the neuroendocrine system: lessons from developmental biology provide insights into human disease. Endocr Rev 2001; 22:2-35. [PMID: 11159814 DOI: 10.1210/edrv.22.1.0421] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
POU domain factors are transcriptional regulators characterized by a highly conserved DNA-binding domain referred to as the POU domain. The structure of the POU domain has been solved, facilitating the understanding of how these proteins bind to DNA and regulate transcription via complex protein-protein interactions. Several members of the POU domain family have been implicated in the control of development and function of the neuroendocrine system. Such roles have been most clearly established for Pit-1, which is required for formation of somatotropes, lactotropes, and thyrotropes in the anterior pituitary gland, and for Brn-2, which is critical for formation of magnocellular and parvocellular neurons in the paraventricular and supraoptic nuclei of the hypothalamus. While genetic evidence is lacking, molecular biology experiments have implicated several other POU factors in the regulation of gene expression in the hypothalamus and pituitary gland. Pit-1 mutations in humans cause combined pituitary hormone deficiency similar to that found in mice deleted for the Pit-1 gene, providing a striking example of how basic developmental biology studies have provided important insights into human disease.
Collapse
Affiliation(s)
- B Andersen
- Department of Medicine, University of California, San Diego, La Jolla, 92093-0648, USA.
| | | |
Collapse
|
41
|
Harvey S, Azumaya Y, Hull KL. Pituitary and extrapituitary growth hormone: Pit-1 dependence? Can J Physiol Pharmacol 2000. [DOI: 10.1139/y00-095] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Growth hormone (GH) is primarily produced in pituitary somatotrophs. The synthesis of this hormone is thought to be dependent upon a pituitary-specific transcription factor (Pit-1). However, many extrapituitary tissues are now known to express GH genes. The extrapituitary production of GH may therefore indicate an extrapituitary distribution of the Pit-1 gene. The extrapituitary production of GH may, alternatively, indicate that GH expression occurs independently of Pit-1 in extrapituitary tissues. These possibilities are considered in this brief review.Key words: growth hormone, pituitary, pituitary transcription factor 1.
Collapse
|
42
|
Marshak S, Benshushan E, Shoshkes M, Havin L, Cerasi E, Melloul D. Functional conservation of regulatory elements in the pdx-1 gene: PDX-1 and hepatocyte nuclear factor 3beta transcription factors mediate beta-cell-specific expression. Mol Cell Biol 2000; 20:7583-90. [PMID: 11003654 PMCID: PMC86312 DOI: 10.1128/mcb.20.20.7583-7590.2000] [Citation(s) in RCA: 94] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The PDX-1 transcription factor plays a key role in pancreatic development and in the regulation of the insulin gene in the adult beta cell. As its functions appear to be similar in humans and mice, we analyzed the functional conservation of homologous sequences important for the maintenance and the cell-specific regulation of the pdx-1 gene. Apart from the proximal promoter region, three highly homologous (PH1 to PH3) sequences were apparent in the human and mouse 5' flanking regions of the gene. By transient transfections in beta and non-beta cells, we show that mainly PH1 and PH2 preferentially confer beta-cell-specific activation on a heterologous promoter. DNase I footprinting and binding analyses revealed that both bind to and are transactivated by hepatocyte nuclear factor 3beta (HNF-3beta). Furthermore, the PH1 enhancer element also binds the PDX-1 transcription factor itself, which acts cooperatively with adjacent HNF-3beta to regulate its transcriptional potency. This finding suggests a possible autoregulatory loop as a mechanism for PDX-1 to control its own expression.
Collapse
Affiliation(s)
- S Marshak
- Department of Endocrinology & Metabolism, Hadassah University Hospital, 91120 Jerusalem, Israel
| | | | | | | | | | | |
Collapse
|
43
|
Mandemakers W, Zwart R, Kraay R, Grosveld G, Jaegle AG, Broos L, Meijer D. Transcriptional regulation of the POU gene Oct-6 in Schwann cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2000; 468:13-22. [PMID: 10635016 DOI: 10.1007/978-1-4615-4685-6_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Genetic evidence suggests that the POU transcription factor Oct-6 plays a pivotal role as an intracellular regulator of Schwann cell differentiation. In the absence of Oct-6 function Schwann cells are generated in appropriate numbers and these cells differentiate normally up to the promyelin stage at which they transiently arrest. During peripheral nerve development Oct-6 expression is initiated in Schwann cell precursors and is strongly upregulated in promyelin cells. Oct-6 expression is subsequently extinguished in terminally differentiating Schwann cells. Thus, identification and characterisation of the DNA elements involved in this stage specific regulation may lead us to the signaling cascade and the axon-derived signals that drive Schwann cell differentiation and initiate myelination. Here we present experiments that aim at identifying such regulatory sequences.
Collapse
Affiliation(s)
- W Mandemakers
- Department of Cell Biology and Genetics, Erasmus University, Rotterdam, Netherlands
| | | | | | | | | | | | | |
Collapse
|
44
|
McEvilly RJ, Rosenfeld MG. The role of POU domain proteins in the regulation of mammalian pituitary and nervous system development. PROGRESS IN NUCLEIC ACID RESEARCH AND MOLECULAR BIOLOGY 1999; 63:223-55. [PMID: 10506833 DOI: 10.1016/s0079-6603(08)60724-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
POU domain proteins represent a subfamily of homeodomain-containing transcription factors that are expressed in many animal orders in a number of distinct regions in the developing and adult organism. In mammals, the expression profiles of these factors have suggested roles for class I, class III, and class IV POU domain proteins in the development, maintenance, and function of the endocrine and nervous systems. The genetic characterizations of the functions of these proteins during the generation, differentiation, and maturation of cells comprising these tissues have revealed a requirement for the individual actions of these transcription factors in the development of various elements of the anterior pituitary, the brain, and the somatosensory, vestibular/cochlear, and visual systems.
Collapse
Affiliation(s)
- R J McEvilly
- Department of Medicine, University of California, San Diego, La Jolla 92093, USA
| | | |
Collapse
|
45
|
Dasen JS, Rosenfeld MG. Combinatorial codes in signaling and synergy: lessons from pituitary development. Curr Opin Genet Dev 1999; 9:566-74. [PMID: 10508698 DOI: 10.1016/s0959-437x(99)00015-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The development of the hormone-secreting cell types in the pituitary gland provides an excellent model system in which to explore the complex transcriptional mechanisms underlying the specification and maintenance of differentiated cell types in mammalian organogenesis. Pituitary development is orchestrated through the combinatorial actions of a repertoire of signaling-gradient-induced transcription factors which, on the basis of their distinct and overlapping expression patterns, and functional interactions, ultimately has led to the generation of functionally distinct cell phenotypes from a common ectodermal primordium.
Collapse
Affiliation(s)
- J S Dasen
- Howard Hughes Medical Institute Cellular and Molecular Medicine University of California San Diego, Department and School of Medicine La Jolla, California, 92093-0648, USA.
| | | |
Collapse
|
46
|
Abstract
The POU-IV or Brn-3 class of transcription factors exhibit conserved structure, DNA-binding properties, and expression in specific subclasses of neurons across widely diverged species. In the mouse CNS, Brn-3.0 expression characterizes specific neurons from neurogenesis through the life of the cell. This irreversible activation of expression suggests positive autoregulation. To search for cis-acting elements that could mediate autoregulation we used a novel method, complex stability screening, which we applied to rapidly identify functional Brn-3.0 recognition sites within a large genomic region encompassing the mouse brn-3.0 locus. This method is based on the observation that the kinetic stability of Brn-3.0 complexes with specific DNA sequences, as measured by their dissociation half-lives, is highly correlated with the ability of those sequences to mediate transcriptional activation by Brn-3.0. The principal Brn-3.0 autoregulatory region lies approximately 5 kb upstream from the Brn-3.0 transcription start site and contains multiple Brn-3.0-binding sites that strongly resemble the optimal binding site for this protein class. This region also mediates transactivation by the closely related protein Brn-3.2, suggesting a regulatory cascade of POU proteins in specific neurons in which Brn-3.2 expression precedes Brn-3.0.
Collapse
|
47
|
Dasen JS, O'Connell SM, Flynn SE, Treier M, Gleiberman AS, Szeto DP, Hooshmand F, Aggarwal AK, Rosenfeld MG. Reciprocal interactions of Pit1 and GATA2 mediate signaling gradient-induced determination of pituitary cell types. Cell 1999; 97:587-98. [PMID: 10367888 DOI: 10.1016/s0092-8674(00)80770-9] [Citation(s) in RCA: 255] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The mechanisms by which transient gradients of signaling molecules lead to emergence of specific cell types remain a central question in mammalian organogenesis. Here, we demonstrate that the appearance of four ventral pituitary cell types is mediated via the reciprocal interactions of two transcription factors, Pit1 and GATA2, which are epistatic to the remainder of the cell type-specific transcription programs and serve as the molecular memory of the transient signaling events. Unexpectedly, this program includes a DNA binding-independent function of Pit1, suppressing the ventral GATA2-dependent gonadotrope program by inhibiting GATA2 binding to gonadotrope- but not thyrotrope-specific genes, indicating that both DNA binding-dependent and -independent actions of abundant determining factors contribute to generate distinct cell phenotypes.
Collapse
Affiliation(s)
- J S Dasen
- Howard Hughes Medical Institute, Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla 92093-0648, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Schonemann MD, Ryan AK, Erkman L, McEvilly RJ, Bermingham J, Rosenfeld MG. POU domain factors in neural development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1999; 449:39-53. [PMID: 10026784 DOI: 10.1007/978-1-4615-4871-3_4] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Transcription factors serve critical roles in the progressive development of general body plan, organ commitment, and finally, specific cell types. Comparison of the biological roles of a series of individual members within a family permits some generalizations to be made regarding the developmental events that are likely to be regulated by a particular class of transcription factors. Here, we evidence that the developmental functions of the family of transcription factors characterized by the POU DNA binding motif exerts roles in mammalian development. The POU domain family of transcription factors was defined following the observation that the products of three mammalian genes, Pit-1, Oct-1, and Oct-2, and the protein encoded by the C. elegans gene unc-86, shared a region of homology, known as the POU domain. The POU domain is a bipartite DNA binding domain, consisting of two highly conserved regions, tethered by a variable linker. The approximately 75 amino acid N-terminal region was called the POU-specific domain and the C-terminal 60 amino acid region, the POU-homeodomain. High-affinity site-specific DNA binding by POU domain transcription factors requires both the POU-specific and the POU-homeodomain. Resolution of the crystal structures of Oct-1 and Pit-1 POU domains bound to DNA as a monomer and homodimer, respectively, confirmed several of the in vitro findings regarding interactions of this bipartite DNA binding domain with DNA and has provided important information regarding the flexibility and versatility of POU domain proteins. Overall the crystal structure of a monomer of the Oct-1 POU domain bound to the octamer element was similar to that predicted by the NMR solution structures of the POU-specific domain and the POU-homeodomain in isolation, with the POU-specific domain consists of four alpha helices, with the second and third helices forming a structure similar to the helix-turn-helix motif of the lambda and 434 repressors; several of the DNA base contacts are also conserved. A homodimer of the Pit-1 POU domain was crystallized bound to a Pit-1 dimer DNA element that is closely related to a site in the proximal promoter of the prolactin gene. The structure of the Pit-1 POU domain on DNA is very similar to that of Oct-1, and the Pit-1 POU-homeodomain/DNA structure is strikingly similar to that of other homeodomains, including the Oct-1 POU-homeodomain. The DNA contacts made by the Pit-1 POU-specific domain are also similar to those of Oct-1 and conserved with many made by the prokaryotic repressors. In the Oct-1 crystal, the POU-specific domain recognizes a GCAT half-site, while the corresponding sequence recognized by the Pit-1 POU-specific domain, GTAT, is on the opposing strand. As a result, the orientation of the Pit-1 POU-specific domain relative to the POU-homeodomain is flipped, as compared to the Oct-1 crystal structure, indicating the remarkable flexibility of the POU-specific domain in adapting to variations in sequence within the site. Also in contrast to the Oct-1 monomer structure is the observation that the POU-specific and POU-homeodomain of each Pit-1 molecule make major groove contacts on the same face of the DNA, consistent with the constraints imposed by its 15 amino acid linker. As a result, the Pit-1 POU domain homodimer essentially surrounds its DNA binding site. In the Pit-1 POU domain homodimer the dimerization interface is formed between the C-terminal end of helix 3 of the POU-homeodomain of one Pit-1 molecule and the N-terminus of helix 1 and the loop between helices 3 and 4 of the POU-specific domain of the other Pit-1 molecule. In contrast to other homeodomain crystal structures, the C-terminus of helix 3 in the Pit-1 POU-homeo-domain has an extended structure. (ABSTRACT TRUNCATED)
Collapse
Affiliation(s)
- M D Schonemann
- Howard Hughes Medical Institute, Department and School of Medicine, University of California, San Diego 92093-0648, USA
| | | | | | | | | | | |
Collapse
|
49
|
Miller AA, Bernardoni R, Giangrande A. Positive autoregulation of the glial promoting factor glide/gcm. EMBO J 1998; 17:6316-26. [PMID: 9799239 PMCID: PMC1170956 DOI: 10.1093/emboj/17.21.6316] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Fly gliogenesis depends on the glial-cell-deficient/glial-cell-missing (glide/gcm) transcription factor. glide/gcm expression is necessary and sufficient to induce the glial fate within and outside the nervous system, indicating that the activity of this gene must be tightly regulated. The current model is that glide/gcm activates the glial fate by inducing the expression of glial-specific genes that are required to maintain such a fate. Previous observations on the null glide/gcmN7-4 allele evoked the possibility that another role of glide/gcm might be to maintain and/or amplify its own expression. Here we show that glide/gcm does positively autoregulate in vitro and in vivo, and that the glide/gcmN7-4 protein is not able to do so. We thereby provide the first direct evidence of both a target and a regulator of glide/gcm. Our data also demonstrate that glide/gcm transcription is regulated at two distinct steps: initiation, which is glide/gcm-independent, and maintenance, which requires glide/gcm. Interestingly, we have found that autoregulation requires the activity of additional cell-specific cofactors. The present results suggest transcriptional autoregulation is a mechanism for glial fate induction.
Collapse
Affiliation(s)
- A A Miller
- Institut de Génétique et Biologie Moléculaire et Cellulaire, IGBMC/CNRS/INSERM/ULP, BP 163 67404 Illkirch, Communauté Urbaine de Strasbourg, France
| | | | | |
Collapse
|
50
|
Ryan AK, Rosenfeld MG. POU domain family values: flexibility, partnerships, and developmental codes. Genes Dev 1997; 11:1207-25. [PMID: 9171367 DOI: 10.1101/gad.11.10.1207] [Citation(s) in RCA: 404] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- A K Ryan
- Howard Hughes Medical Institute, Department and School of Medicine, University of California at San Diego, La Jolla 92093-0648, USA
| | | |
Collapse
|