1
|
Rizzoti K, Chakravarty P, Sheridan D, Lovell-Badge R. SOX9-positive pituitary stem cells differ according to their position in the gland and maintenance of their progeny depends on context. SCIENCE ADVANCES 2023; 9:eadf6911. [PMID: 37792947 PMCID: PMC10550238 DOI: 10.1126/sciadv.adf6911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 09/01/2023] [Indexed: 10/06/2023]
Abstract
Stem cell (SC) differentiation and maintenance of resultant progeny underlie cell turnover in many organs, but it is difficult to pinpoint the contribution of either process. In the pituitary, a central regulator of endocrine axes, adult SCs undergo activation after target organ ablation, providing a well-characterized paradigm to study an adaptative response in a multi-organ system. Here, we used single-cell technologies to characterize SC heterogeneity and mobilization together with lineage tracing. We show that SC differentiation occurs more frequently than thought previously. In adaptative conditions, differentiation increases and is more diverse than demonstrated by the lineage tracing experiments. Detailed examination of SC progeny suggests that maintenance of selected nascent cells underlies SC output, highlighting a trophic role for the microenvironment. Analyses of cell trajectories further predict pathways and potential regulators. Our model provides a valuable system to study the influence of evolving states on the mechanisms of SC mobilization.
Collapse
Affiliation(s)
- Karine Rizzoti
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London NW1 1AT, UK
| | | | - Daniel Sheridan
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London NW1 1AT, UK
| | - Robin Lovell-Badge
- Laboratory of Stem Cell Biology and Developmental Genetics, The Francis Crick Institute, London NW1 1AT, UK
| |
Collapse
|
2
|
Scagliotti V, Vignola ML, Willis T, Howard M, Marinelli E, Gaston-Massuet C, Andoniadou C, Charalambous M. Imprinted Dlk1 dosage as a size determinant of the mammalian pituitary gland. eLife 2023; 12:e84092. [PMID: 37589451 PMCID: PMC10468206 DOI: 10.7554/elife.84092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 08/16/2023] [Indexed: 08/18/2023] Open
Abstract
Co-regulated genes of the Imprinted Gene Network are involved in the control of growth and body size, and imprinted gene dysfunction underlies human paediatric disorders involving the endocrine system. Imprinted genes are highly expressed in the pituitary gland, among them, Dlk1, a paternally expressed gene whose membrane-bound and secreted protein products can regulate proliferation and differentiation of multiple stem cell populations. Dosage of circulating DLK1 has been previously implicated in the control of growth through unknown molecular mechanisms. Here we generate a series of mouse genetic models to modify levels of Dlk1 expression in the pituitary gland and demonstrate that the dosage of DLK1 modulates the process of stem cell commitment with lifelong impact on pituitary gland size. We establish that stem cells are a critical source of DLK1, where embryonic disruption alters proliferation in the anterior pituitary, leading to long-lasting consequences on growth hormone secretion later in life.
Collapse
Affiliation(s)
- Valeria Scagliotti
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King’s College LondonLondonUnited Kingdom
| | - Maria Lillina Vignola
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King’s College LondonLondonUnited Kingdom
| | - Thea Willis
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King’s College LondonLondonUnited Kingdom
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College LondonLondonUnited Kingdom
| | - Mark Howard
- MRC Centre for Transplantation, Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King’s College LondonLondonUnited Kingdom
| | - Eugenia Marinelli
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King’s College LondonLondonUnited Kingdom
| | - Carles Gaston-Massuet
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of LondonLondonUnited Kingdom
| | - Cynthia Andoniadou
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College LondonLondonUnited Kingdom
| | - Marika Charalambous
- Department of Medical and Molecular Genetics, Faculty of Life Sciences and Medicine, King’s College LondonLondonUnited Kingdom
| |
Collapse
|
3
|
Vamvoukaki R, Chrysoulaki M, Betsi G, Xekouki P. Pituitary Tumorigenesis-Implications for Management. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59040812. [PMID: 37109772 PMCID: PMC10145673 DOI: 10.3390/medicina59040812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023]
Abstract
Pituitary neuroendocrine tumors (PitNETs), the third most common intracranial tumor, are mostly benign. However, some of them may display a more aggressive behavior, invading into the surrounding structures. While they may rarely metastasize, they may resist different treatment modalities. Several major advances in molecular biology in the past few years led to the discovery of the possible mechanisms involved in pituitary tumorigenesis with a possible therapeutic implication. The mutations in the different proteins involved in the Gsa/protein kinase A/c AMP signaling pathway are well-known and are responsible for many PitNETS, such as somatotropinomas and, in the context of syndromes, as the McCune-Albright syndrome, Carney complex, familiar isolated pituitary adenoma (FIPA), and X-linked acrogigantism (XLAG). The other pathways involved are the MAPK/ERK, PI3K/Akt, Wnt, and the most recently studied HIPPO pathways. Moreover, the mutations in several other tumor suppressor genes, such as menin and CDKN1B, are responsible for the MEN1 and MEN4 syndromes and succinate dehydrogenase (SDHx) in the context of the 3PAs syndrome. Furthermore, the pituitary stem cells and miRNAs hold an essential role in pituitary tumorigenesis and may represent new molecular targets for their diagnosis and treatment. This review aims to summarize the different cell signaling pathways and genes involved in pituitary tumorigenesis in an attempt to clarify their implications for diagnosis and management.
Collapse
Affiliation(s)
- Rodanthi Vamvoukaki
- Endocrinology and Diabetes Clinic, University Hospital of Heraklion, School of Medicine, University of Crete, 71500 Crete, Greece
| | - Maria Chrysoulaki
- Endocrinology and Diabetes Clinic, University Hospital of Heraklion, School of Medicine, University of Crete, 71500 Crete, Greece
| | - Grigoria Betsi
- Endocrinology and Diabetes Clinic, University Hospital of Heraklion, School of Medicine, University of Crete, 71500 Crete, Greece
| | - Paraskevi Xekouki
- Endocrinology and Diabetes Clinic, University Hospital of Heraklion, School of Medicine, University of Crete, 71500 Crete, Greece
| |
Collapse
|
4
|
Zhang J, Zhou Y, Guo J, Li L, Liu H, Lu C, Jiang Y, Cui S. MicroRNA-7a2 is required for the development of pituitary stem cells. Stem Cells Dev 2022; 31:357-368. [PMID: 35652338 DOI: 10.1089/scd.2022.0023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The pituitary gland is inhabited by a subpopulation of SOX2+ stem cells. However, the regulatory mechanisms underlying pituitary stem cell development remain poorly understood. Here, we demonstrate that microRNA-7a (miR-7a) is enriched in the developing pituitary and is spatiotemporally expressed in the pituitary stem cells. Constitutive deletion of miR-7a2 in mice results in pituitary dysplasia emerging during birth, which is primarily manifested as malformed anterior lobes. Using immunofluorescence, immunohistochemistry or in situ hybridization, we observe that the specification of hormone-expressing cells is not impeded post miR-7a2 deletion at birth, although the terminal differentiation of gonadotropes is inhibited. Further investigation of neonatal and adult pituitaries in miR-7a2 knockout mice reveals an expansion of the SOX2+ pituitary stem cell compartment. The inhibition of epithelial-mesenchymal like transition seems to be responsible for this phenotype, rather than abnormal proliferation or apoptosis. Furthermore, our data suggest that Gli3 and Ckap4 are potential targets of miR-7a in pituitary stem cells. In summary, our results identify miR-7a2 as a crucial factor involved in pituitary stem cell development.
Collapse
Affiliation(s)
- Jinglin Zhang
- Yangzhou University, 38043, Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou, Jiangsu, China.,Yangzhou University, 38043, Institute of Reproduction and Metabolism, Yangzhou, Jiangsu, China;
| | - Yewen Zhou
- Yangzhou University, 38043, College of Veterinary Medicine, Yangzhou, Jiangsu, China.,Yangzhou University, 38043, Institute of Reproduction and Metabolism, Yangzhou, Jiangsu, China;
| | - Jiajia Guo
- Yangzhou University, 38043, College of Veterinary Medicine, Yangzhou, Jiangsu, China;
| | - Liuhui Li
- Yangzhou University, 38043, College of Veterinary Medicine, Yangzhou, Jiangsu, China;
| | - Hui Liu
- Yangzhou University, 38043, College of Veterinary Medicine, Yangzhou, Jiangsu, China;
| | - Chenyang Lu
- Yangzhou University, 38043, College of Veterinary Medicine, Yangzhou, Jiangsu, China;
| | - Ying Jiang
- Yangzhou University, 38043, College of Veterinary Medicine, Yangzhou, Jiangsu, China;
| | - Sheng Cui
- Yangzhou University, 38043, College of Veterinary Medicine, Yangzhou, Jiangsu, China.,Yangzhou University, 38043, Institute of Reproduction and Metabolism, Yangzhou, Jiangsu, China.,Yangzhou University, 38043, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu, China;
| |
Collapse
|
5
|
Guérineau NC, Campos P, Le Tissier PR, Hodson DJ, Mollard P. Cell Networks in Endocrine/Neuroendocrine Gland Function. Compr Physiol 2022; 12:3371-3415. [PMID: 35578964 DOI: 10.1002/cphy.c210031] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Reproduction, growth, stress, and metabolism are determined by endocrine/neuroendocrine systems that regulate circulating hormone concentrations. All these systems generate rhythms and changes in hormone pulsatility observed in a variety of pathophysiological states. Thus, the output of endocrine/neuroendocrine systems must be regulated within a narrow window of effective hormone concentrations but must also maintain a capacity for plasticity to respond to changing physiological demands. Remarkably most endocrinologists still have a "textbook" view of endocrine gland organization which has emanated from 20th century histological studies on thin 2D tissue sections. However, 21st -century technological advances, including in-depth 3D imaging of specific cell types have vastly changed our knowledge. We now know that various levels of multicellular organization can be found across different glands, that organizational motifs can vary between species and can be modified to enhance or decrease hormonal release. This article focuses on how the organization of cells regulates hormone output using three endocrine/neuroendocrine glands that present different levels of organization and complexity: the adrenal medulla, with a single neuroendocrine cell type; the anterior pituitary, with multiple intermingled cell types; and the pancreas with multiple intermingled cell types organized into distinct functional units. We give an overview of recent methodologies that allow the study of the different components within endocrine systems, particularly their temporal and spatial relationships. We believe the emerging findings about network organization, and its impact on hormone secretion, are crucial to understanding how homeostatic regulation of endocrine axes is carried out within endocrine organs themselves. © 2022 American Physiological Society. Compr Physiol 12:3371-3415, 2022.
Collapse
Affiliation(s)
| | - Pauline Campos
- College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, UK
| | - Paul R Le Tissier
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, Scotland, UK
| | - David J Hodson
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Edgbaston, UK.,Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK.,COMPARE University of Birmingham and University of Nottingham Midlands, UK.,Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), NIHR Oxford Biomedical Research Centre, Churchill Hospital, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Patrice Mollard
- IGF, University of Montpellier, CNRS, INSERM, Montpellier, France
| |
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
|
Russell JP, Lim X, Santambrogio A, Yianni V, Kemkem Y, Wang B, Fish M, Haston S, Grabek A, Hallang S, Lodge EJ, Patist AL, Schedl A, Mollard P, Nusse R, Andoniadou CL. Pituitary stem cells produce paracrine WNT signals to control the expansion of their descendant progenitor cells. eLife 2021; 10:59142. [PMID: 33399538 PMCID: PMC7803373 DOI: 10.7554/elife.59142] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 01/04/2021] [Indexed: 12/16/2022] Open
Abstract
In response to physiological demand, the pituitary gland generates new hormone-secreting cells from committed progenitor cells throughout life. It remains unclear to what extent pituitary stem cells (PSCs), which uniquely express SOX2, contribute to pituitary growth and renewal. Moreover, neither the signals that drive proliferation nor their sources have been elucidated. We have used genetic approaches in the mouse, showing that the WNT pathway is essential for proliferation of all lineages in the gland. We reveal that SOX2+ stem cells are a key source of WNT ligands. By blocking secretion of WNTs from SOX2+ PSCs in vivo, we demonstrate that proliferation of neighbouring committed progenitor cells declines, demonstrating that progenitor multiplication depends on the paracrine WNT secretion from SOX2+ PSCs. Our results indicate that stem cells can hold additional roles in tissue expansion and homeostasis, acting as paracrine signalling centres to coordinate the proliferation of neighbouring cells.
Collapse
Affiliation(s)
- John P Russell
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Xinhong Lim
- Skin Research Institute of Singapore, Agency for Science, Technology and Research, Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Alice Santambrogio
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom.,Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Val Yianni
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Yasmine Kemkem
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Montpellier, France
| | - Bruce Wang
- Howard Hughes Medical Institute, Stanford University School of Medicine, Department of Developmental Biology, Stanford University School of Medicine, Stanford, United States.,Department of Medicine and Liver Center, University of California San Francisco, San Francisco, United States
| | - Matthew Fish
- Howard Hughes Medical Institute, Stanford University School of Medicine, Department of Developmental Biology, Stanford University School of Medicine, Stanford, United States
| | - Scott Haston
- Developmental Biology and Cancer, Birth Defects Research Centre, UCL GOS Institute of Child Health, London, United Kingdom
| | | | - Shirleen Hallang
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Emily J Lodge
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Amanda L Patist
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | | | - Patrice Mollard
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, Montpellier, France
| | - Roel Nusse
- Howard Hughes Medical Institute, Stanford University School of Medicine, Department of Developmental Biology, Stanford University School of Medicine, Stanford, United States
| | - Cynthia L Andoniadou
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom.,Department of Medicine III, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
8
|
Zhang S, Cui Y, Ma X, Yong J, Yan L, Yang M, Ren J, Tang F, Wen L, Qiao J. Single-cell transcriptomics identifies divergent developmental lineage trajectories during human pituitary development. Nat Commun 2020; 11:5275. [PMID: 33077725 PMCID: PMC7572359 DOI: 10.1038/s41467-020-19012-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 09/23/2020] [Indexed: 12/01/2022] Open
Abstract
The anterior pituitary gland plays a central role in regulating various physiological processes, including body growth, reproduction, metabolism and stress response. Here, we perform single-cell RNA-sequencing (scRNA-seq) of 4113 individual cells from human fetal pituitaries. We characterize divergent developmental trajectories with distinct transitional intermediate states in five hormone-producing cell lineages. Corticotropes exhibit an early intermediate state prior to full differentiation. Three cell types of the PIT-1 lineage (somatotropes, lactotropes and thyrotropes) segregate from a common progenitor coexpressing lineage-specific transcription factors of different sublineages. Gonadotropes experience two multistep developmental trajectories. Furthermore, we identify a fetal gonadotrope cell subtype expressing the primate-specific hormone chorionic gonadotropin. We also characterize the cellular heterogeneity of pituitary stem cells and identify a hybrid epithelial/mesenchymal state and an early-to-late state transition. Here, our results provide insights into the transcriptional landscape of human pituitary development, defining distinct cell substates and subtypes and illustrating transcription factor dynamics during cell fate commitment. Editor’s summary_NCOMMS-19-41732B The anterior pituitary gland controls body growth and reproduction but how early development is dynamically regulated is unclear. Here, the authors use scRNA-seq of human fetal pituitaries to identify different developmental routes and state transitions of five hormone-producing cell lineages, and a hybrid epithelial/mesenchymal state of pituitary stem cells.
Collapse
Affiliation(s)
- Shu Zhang
- Department of Obstetrics and Gynecology, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Third Hospital, Peking University, Beijing, 100871, China.,Biomedical Pioneering Innovation Center, School of Life Sciences, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, 100871, China
| | - Yueli Cui
- Department of Obstetrics and Gynecology, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Third Hospital, Peking University, Beijing, 100871, China.,Biomedical Pioneering Innovation Center, School of Life Sciences, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, 100871, China
| | - Xinyi Ma
- Department of Obstetrics and Gynecology, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Third Hospital, Peking University, Beijing, 100871, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
| | - Jun Yong
- Department of Obstetrics and Gynecology, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Third Hospital, Peking University, Beijing, 100871, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
| | - Liying Yan
- Department of Obstetrics and Gynecology, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Third Hospital, Peking University, Beijing, 100871, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
| | - Ming Yang
- Department of Obstetrics and Gynecology, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Third Hospital, Peking University, Beijing, 100871, China.,Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China.,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China.,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Jie Ren
- Department of Obstetrics and Gynecology, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Third Hospital, Peking University, Beijing, 100871, China.,Biomedical Pioneering Innovation Center, School of Life Sciences, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, 100871, China
| | - Fuchou Tang
- Department of Obstetrics and Gynecology, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Third Hospital, Peking University, Beijing, 100871, China.,Biomedical Pioneering Innovation Center, School of Life Sciences, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, 100871, China.,Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Lu Wen
- Department of Obstetrics and Gynecology, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Third Hospital, Peking University, Beijing, 100871, China. .,Biomedical Pioneering Innovation Center, School of Life Sciences, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, 100871, China.
| | - Jie Qiao
- Department of Obstetrics and Gynecology, Beijing Advanced Innovation Center for Genomics, School of Life Sciences, Third Hospital, Peking University, Beijing, 100871, China. .,Biomedical Pioneering Innovation Center, School of Life Sciences, Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, 100871, China. .,Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China. .,Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China.
| |
Collapse
|
9
|
O'Hara L, Christian HC, Jeffery N, Le Tissier P, Smith LB. Characterisation of a mural cell network in the murine pituitary gland. J Neuroendocrinol 2020; 32:e12903. [PMID: 32959418 DOI: 10.1111/jne.12903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 08/03/2020] [Accepted: 08/21/2020] [Indexed: 11/29/2022]
Abstract
The anterior and intermediate lobes of the pituitary are composed of endocrine cells, as well as vasculature and supporting cells, such as folliculostellate cells. Folliculostellate cells form a network with several postulated roles in the pituitary, including production of paracrine signalling molecules and cytokines, coordination of endocrine cell hormone release, phagocytosis, and structural support. Folliculostellate cells in rats are characterised by expression of S100B protein, and in humans by glial fibrillary acid protein. However, there is evidence for another network of supporting cells in the anterior pituitary that has properties of mural cells, such as vascular smooth muscle cells and pericytes. The present study aims to characterise the distribution of cells that express the mural cell marker platelet derived growth factor receptor beta (PDGFRβ) in the mouse pituitary and establish whether these cells are folliculostellate. By immunohistochemical localisation, we determine that approximately 80% of PDGFRβ+ cells in the mouse pituitary have a non-perivascular location and 20% are pericytes. Investigation of gene expression in a magnetic cell sorted population of PDGFRβ+ cells shows that, despite a mostly non-perivascular location, this population is enriched for mural cell markers but not enriched for rat or human folliculostellate cell markers. This is confirmed by immunohistochemistry. The present study concludes that a mural cell network is present throughout the anterior pituitary of the mouse and that this population does not express well-characterised human or rat folliculostellate cell markers.
Collapse
Affiliation(s)
- Laura O'Hara
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Helen C Christian
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Nathan Jeffery
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Paul Le Tissier
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Lee B Smith
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
- School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
10
|
Abstract
The development of the anterior pituitary gland occurs in distinct sequential developmental steps, leading to the formation of a complex organ containing five different cell types secreting six different hormones. During this process, the temporal and spatial expression of a cascade of signaling molecules and transcription factors plays a crucial role in organ commitment, cell proliferation, patterning, and terminal differentiation. The morphogenesis of the gland and the emergence of distinct cell types from a common primordium are governed by complex regulatory networks involving transcription factors and signaling molecules that may be either intrinsic to the developing pituitary or extrinsic, originating from the ventral diencephalon, the oral ectoderm, and the surrounding mesenchyme. Endocrine cells of the pituitary gland are organized into structural and functional networks that contribute to the coordinated response of endocrine cells to stimuli; these cellular networks are formed during embryonic development and are maintained or may be modified in adulthood, contributing to the plasticity of the gland. Abnormalities in any of the steps of pituitary development may lead to congenital hypopituitarism that includes a spectrum of disorders from isolated to combined hormone deficiencies including syndromic disorders such as septo-optic dysplasia. Over the past decade, the acceleration of next-generation sequencing has allowed for rapid analysis of the patient genome to identify novel mutations and novel candidate genes associated with hypothalmo-pituitary development. Subsequent functional analysis using patient fibroblast cells, and the generation of stem cells derived from patient cells, is fast replacing the need for animal models while providing a more physiologically relevant characterization of novel mutations. Furthermore, CRISPR-Cas9 as the method for gene editing is replacing previous laborious and time-consuming gene editing methods that were commonly used, thus yielding knockout cell lines in a fraction of the time. © 2020 American Physiological Society. Compr Physiol 10:389-413, 2020.
Collapse
Affiliation(s)
- Kyriaki S Alatzoglou
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London (UCL), London, UK
| | - Louise C Gregory
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London (UCL), London, UK
| | - Mehul T Dattani
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, University College London (UCL), London, UK
| |
Collapse
|
11
|
Mariniello K, Ruiz-Babot G, McGaugh EC, Nicholson JG, Gualtieri A, Gaston-Massuet C, Nostro MC, Guasti L. Stem Cells, Self-Renewal, and Lineage Commitment in the Endocrine System. Front Endocrinol (Lausanne) 2019; 10:772. [PMID: 31781041 PMCID: PMC6856655 DOI: 10.3389/fendo.2019.00772] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 10/23/2019] [Indexed: 12/15/2022] Open
Abstract
The endocrine system coordinates a wide array of body functions mainly through secretion of hormones and their actions on target tissues. Over the last decades, a collective effort between developmental biologists, geneticists, and stem cell biologists has generated a wealth of knowledge related to the contribution of stem/progenitor cells to both organogenesis and self-renewal of endocrine organs. This review provides an up-to-date and comprehensive overview of the role of tissue stem cells in the development and self-renewal of endocrine organs. Pathways governing crucial steps in both development and stemness maintenance, and that are known to be frequently altered in a wide array of endocrine disorders, including cancer, are also described. Crucially, this plethora of information is being channeled into the development of potential new cell-based treatment modalities for endocrine-related illnesses, some of which have made it through clinical trials.
Collapse
Affiliation(s)
- Katia Mariniello
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Gerard Ruiz-Babot
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, United States
- Harvard Stem Cell Institute, Cambridge, MA, United States
| | - Emily C. McGaugh
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - James G. Nicholson
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Angelica Gualtieri
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Carles Gaston-Massuet
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Maria Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Leonardo Guasti
- Centre for Endocrinology, William Harvey Research Institute, Bart's and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
12
|
Anastassiadis C, Jones SL, Pruessner JC. Imaging the pituitary in psychopathologies: a review of in vivo magnetic resonance imaging studies. Brain Struct Funct 2019; 224:2587-2601. [DOI: 10.1007/s00429-019-01942-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 08/13/2019] [Indexed: 12/17/2022]
|
13
|
Montenegro YHA, de Queiroga Nascimento D, de Assis TO, Santos-Lopes SSD. The epigenetics of the hypothalamic-pituitary-adrenal axis in fetal development. Ann Hum Genet 2019; 83:195-213. [PMID: 30843189 DOI: 10.1111/ahg.12306] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 02/13/2019] [Accepted: 02/14/2019] [Indexed: 01/16/2023]
Abstract
The hypothalamic-pituitary-adrenal (HPA) axis is an important hormonal mechanism of the human body and is extremely programmable during embryonic and fetal development. Analyzing its development in this period is the key to understanding in fact how vulnerabilities of congenital diseases occur and any other changes in the phenotypic and histophysiological aspects of the fetus. The environment in which the mother is exposed during the gestational period can influence this axis. Knowing this, our objective was to analyze in recent research the possible impact of epigenetic programming on the HPA axis and its consequences for fetal development. This review brought together articles from two databases: ScienceDirect and PUBMED researched based on key words such as "epigenetics, HPA axis, cardiovascular disease, and circulatory problems" where it demonstrated full relevance in experimental and scientific settings. A total of 101 articles were selected following the criteria established by the researchers. Thus, it was possible to verify that the development of the HPA axis is directly related to changes that occur in the cardiovascular system, to the cerebral growth and other systems depending on the influence that it receives in the period of fetal formation.
Collapse
|
14
|
Bjelobaba I, Janjic MM, Prévide RM, Abebe D, Kucka M, Stojilkovic SS. Distinct Expression Patterns of Osteopontin and Dentin Matrix Protein 1 Genes in Pituitary Gonadotrophs. Front Endocrinol (Lausanne) 2019; 10:248. [PMID: 31057484 PMCID: PMC6478748 DOI: 10.3389/fendo.2019.00248] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 03/29/2019] [Indexed: 01/06/2023] Open
Abstract
Cell-matrix interactions play important roles in pituitary development, physiology, and pathogenesis. In other tissues, a family of non-collagenous proteins, termed SIBLINGs, are known to contribute to cell-matrix interactions. Anterior pituitary gland expresses two SIBLING genes, Dmp1 (dentin matrix protein-1) and Spp1 (secreted phosphoprotein-1) encoding DMP1 and osteopontin proteins, respectively, but their expression pattern and roles in pituitary functions have not been clarified. Here we provide novel evidence supporting the conclusion that Spp1/osteopontin, like Dmp1/DMP1, are expressed in gonadotrophs in a sex- and age-specific manner. Other anterior pituitary cell types do not express these genes. In contrast to Dmp1, Spp1 expression is higher in males; in females, the expression reaches the peak during the diestrus phase of estrous cycle. In further contrast to Dmp1 and marker genes for gonadotrophs, the expression of Spp1 is not regulated by gonadotropin-releasing hormone in vivo and in vitro. However, Spp1 expression increases progressively after pituitary cell dispersion in both female and male cultures. We may speculate that gonadotrophs signal to other pituitary cell types about changes in the structure of pituitary cell-matrix network by osteopontin, a function consistent with the role of this secretory protein in postnatal tissue remodeling, extracellular matrix reorganization after injury, and tumorigenesis.
Collapse
Affiliation(s)
- Ivana Bjelobaba
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
- Institute for Biological Research Sinisa Stankovic, University of Belgrade, Belgrade, Serbia
| | - Marija M. Janjic
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
- Institute for Biological Research Sinisa Stankovic, University of Belgrade, Belgrade, Serbia
| | - Rafael Maso Prévide
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Daniel Abebe
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Marek Kucka
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
| | - Stanko S. Stojilkovic
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health (NIH), Bethesda, MD, United States
- *Correspondence: Stanko S. Stojilkovic
| |
Collapse
|
15
|
Youngblood JL, Coleman TF, Davis SW. Regulation of Pituitary Progenitor Differentiation by β-Catenin. Endocrinology 2018; 159:3287-3305. [PMID: 30085028 DOI: 10.1210/en.2018-00563] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 07/24/2018] [Indexed: 02/06/2023]
Abstract
The pituitary gland is a critical organ that is necessary for many physiological processes, including growth, reproduction, and stress response. The secretion of pituitary hormones from specific cell types regulates these essential processes. Pituitary hormone cell types arise from a common pool of pituitary progenitors, and mutations that disrupt the formation and differentiation of pituitary progenitors result in hypopituitarism. Canonical WNT signaling through CTNNB1 (β-catenin) is known to regulate the formation of the POU1F1 lineage of pituitary cell types. When β-catenin is deleted during the initial formation of the pituitary progenitors, Pou1f1 is not transcribed, which leads to the loss of the POU1F1 lineage. However, when β-catenin is deleted after lineage specification, there is no observable effect. Similarly, the generation of a β-catenin gain-of-function allele in early pituitary progenitors or stem cells results in the formation of craniopharyngiomas, whereas stimulating β-catenin in differentiated cell types has no effect. PROP1 is a pituitary-specific transcription factor, and the peak of PROP1 expression coincides with a critical time point in pituitary organogenesis-that is, after pituitary progenitor formation but before lineage specification. We used a Prop1-cre to conduct both loss- and gain-of-function studies on β-catenin during this critical time point. Our results demonstrate that pituitary progenitors remain sensitive to both loss and gain of β-catenin at this time point, and that either manipulation results in hypopituitarism.
Collapse
Affiliation(s)
- Julie L Youngblood
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina
| | - Tanner F Coleman
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina
| | - Shannon W Davis
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina
| |
Collapse
|
16
|
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
|
17
|
Ellsworth BS, Stallings CE. Molecular Mechanisms Governing Embryonic Differentiation of Pituitary Somatotropes. Trends Endocrinol Metab 2018; 29:510-523. [PMID: 29759686 DOI: 10.1016/j.tem.2018.04.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 04/18/2018] [Accepted: 04/19/2018] [Indexed: 02/07/2023]
Abstract
Pituitary somatotropes secrete growth hormone (GH), which is essential for normal growth and metabolism. Somatotrope defects result in GH deficiency (GHD), leading to short stature in childhood and increased cardiovascular morbidity and mortality in adulthood. Current hormone replacement therapies fail to recapitulate normal pulsatile GH secretion. Stem cell therapies could overcome this problem but are dependent on a thorough understanding of somatotrope differentiation. Although several transcription factors, signaling pathways, and hormones that regulate this process have been identified, the mechanisms of action are not well understood. The purpose of this review is to highlight the known players in somatotrope differentiation while emphasizing the need to better understand these pathways to serve patients with GHD.
Collapse
Affiliation(s)
- Buffy S Ellsworth
- Department of Physiology, School of Medicine, Southern Illinois University, Carbondale, IL 62901-6523, USA.
| | - Caitlin E Stallings
- Department of Physiology, School of Medicine, Southern Illinois University, Carbondale, IL 62901-6523, USA
| |
Collapse
|
18
|
Cheung L, Le Tissier P, Goldsmith SGJ, Treier M, Lovell-Badge R, Rizzoti K. NOTCH activity differentially affects alternative cell fate acquisition and maintenance. eLife 2018; 7:e33318. [PMID: 29578405 PMCID: PMC5889214 DOI: 10.7554/elife.33318] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 03/24/2018] [Indexed: 01/08/2023] Open
Abstract
The pituitary is an essential endocrine gland regulating multiple processes. Regeneration of endocrine cells is of therapeutic interest and recent studies are promising, but mechanisms of endocrine cell fate acquisition need to be better characterised. The NOTCH pathway is important during pituitary development. Here, we further characterise its role in the murine pituitary, revealing differential sensitivity within and between lineages. In progenitors, NOTCH activation blocks cell fate acquisition, with time-dependant modulation. In differentiating cells, response to activation is blunted in the POU1F1 lineage, with apparently normal cell fate specification, while POMC cells remain sensitive. Absence of apparent defects in Pou1f1-Cre; Rbpjfl/fl mice further suggests no direct role for NOTCH signalling in POU1F1 cell fate acquisition. In contrast, in the POMC lineage, NICD expression induces a regression towards a progenitor-like state, suggesting that the NOTCH pathway specifically blocks POMC cell differentiation. These results have implications for pituitary development, plasticity and regeneration. Activation of NOTCH signalling in different cell lineages of the embryonic murine pituitary uncovers an unexpected differential sensitivity, and this consequently reveals new aspects of endocrine lineages development and plasticity.
Collapse
Affiliation(s)
- Leonard Cheung
- Department of Human GeneticsUniversity of MichiganAnn ArborUnited States
| | - Paul Le Tissier
- Centre for Discovery Brain ScienceIntegrative PhysiologyEdinburghUnited Kingdom
| | | | - Mathias Treier
- Cardiovascular and Metabolic SciencesMax Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC)BerlinGermany
- Charité-Universitätsmedizin BerlinBerlinGermany
| | | | | |
Collapse
|
19
|
Fletcher PA, Sherman A, Stojilkovic SS. Common and diverse elements of ion channels and receptors underlying electrical activity in endocrine pituitary cells. Mol Cell Endocrinol 2018; 463:23-36. [PMID: 28652171 PMCID: PMC5742314 DOI: 10.1016/j.mce.2017.06.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 06/20/2017] [Accepted: 06/22/2017] [Indexed: 12/30/2022]
Abstract
The pituitary gland contains six types of endocrine cells defined by hormones they secrete: corticotrophs, melanotrophs, gonadotrophs, thyrotrophs, somatotrophs, and lactotrophs. All these cell types are electrically excitable, and voltage-gated calcium influx is the major trigger for their hormone secretion. Along with hormone intracellular content, G-protein-coupled receptor and ion channel expression can also be considered as defining cell type identity. While many aspects of the developmental and activity dependent regulation of hormone and G-protein-coupled receptor expression have been elucidated, much less is known about the regulation of the ion channels needed for excitation-secretion coupling in these cells. We compare the spontaneous and receptor-controlled patterns of electrical signaling among endocrine pituitary cell types, including insights gained from mathematical modeling. We argue that a common set of ionic currents unites these cells, while differential expression of another subset of ionic currents could underlie cell type-specific patterns. We demonstrate these ideas using a generic mathematical model, showing that it reproduces many observed features of pituitary electrical signaling. Mapping these observations to the developmental lineage suggests possible modes of regulation that may give rise to mature pituitary cell types.
Collapse
Affiliation(s)
- Patrick A Fletcher
- Laboratory of Biological Modeling, National Institute of Diabetes, Digestive and Kidney Diseases, NIH, Bethesda, MD, USA.
| | - Arthur Sherman
- Laboratory of Biological Modeling, National Institute of Diabetes, Digestive and Kidney Diseases, NIH, Bethesda, MD, USA
| | - Stanko S Stojilkovic
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| |
Collapse
|
20
|
Araki T, Liu NA. Cell Cycle Regulators and Lineage-Specific Therapeutic Targets for Cushing Disease. Front Endocrinol (Lausanne) 2018; 9:444. [PMID: 30147673 PMCID: PMC6096271 DOI: 10.3389/fendo.2018.00444] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 07/18/2018] [Indexed: 11/22/2022] Open
Abstract
Cell cycle proteins are critical to pituitary development, but their contribution to lineage-specific tumorigenesis has not been well-elucidated. Emerging evidence from in vitro human tumor analysis and transgenic mouse models indicates that G1/S-related cell cycle proteins, particularly cyclin E, p27, Rb, and E2F1, drive molecular mechanisms that underlie corticotroph-specific differentiation and development of Cushing disease. The aim of this review is to summarize the literature and discuss the complex role of cell cycle regulation in Cushing disease, with a focus on identifying potential targets for therapeutic intervention in patients with these tumors.
Collapse
Affiliation(s)
- Takako Araki
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Minnesota, Minneapolis, MN, United States
- *Correspondence: Takako Araki
| | - Ning-Ai Liu
- Department of Medicine, Pituitary Center, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
21
|
Zimmer B, Piao J, Ramnarine K, Tomishima MJ, Tabar V, Studer L. Derivation of Diverse Hormone-Releasing Pituitary Cells from Human Pluripotent Stem Cells. Stem Cell Reports 2017; 6:858-872. [PMID: 27304916 PMCID: PMC4912387 DOI: 10.1016/j.stemcr.2016.05.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 05/07/2016] [Accepted: 05/10/2016] [Indexed: 12/27/2022] Open
Abstract
Human pluripotent stem cells (hPSCs) provide an unlimited cell source for regenerative medicine. Hormone-producing cells are particularly suitable for cell therapy, and hypopituitarism, a defect in pituitary gland function, represents a promising therapeutic target. Previous studies have derived pituitary lineages from mouse and human ESCs using 3D organoid cultures that mimic the complex events underlying pituitary gland development in vivo. Instead of relying on unknown cellular signals, we present a simple and efficient strategy to derive human pituitary lineages from hPSCs using monolayer culture conditions suitable for cell manufacturing. We demonstrate that purified placode cells can be directed into pituitary fates using defined signals. hPSC-derived pituitary cells show basal and stimulus-induced hormone release in vitro and engraftment and hormone release in vivo after transplantation into a murine model of hypopituitarism. This work lays the foundation for future cell therapy applications in patients with hypopituitarism. Defined, cGMP-ready protocol to derive anterior pituitary-lineage cells from hPSCs FGF8 and BMP2 patterning enables enrichment for specific hormone-producing cells Pituitary cells secrete multiple hormones and respond to physiological stimuli hPSC-pituitary cells partially rescue a rat model of hypopituitarism
Collapse
Affiliation(s)
- Bastian Zimmer
- The Center for Stem Cell Biology, Sloan Kettering Institute for Cancer Research, 1275 York Avenue, New York, NY 10065, USA; Developmental Biology Program, Sloan Kettering Institute for Cancer Research, 1275 York Avenue, New York, NY 10065, USA
| | - Jinghua Piao
- The Center for Stem Cell Biology, Sloan Kettering Institute for Cancer Research, 1275 York Avenue, New York, NY 10065, USA; Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Kiran Ramnarine
- The Center for Stem Cell Biology, Sloan Kettering Institute for Cancer Research, 1275 York Avenue, New York, NY 10065, USA; SKI Stem Cell Research Facility, 1275 York Avenue, New York, NY 10065, USA
| | - Mark J Tomishima
- The Center for Stem Cell Biology, Sloan Kettering Institute for Cancer Research, 1275 York Avenue, New York, NY 10065, USA; SKI Stem Cell Research Facility, 1275 York Avenue, New York, NY 10065, USA
| | - Viviane Tabar
- The Center for Stem Cell Biology, Sloan Kettering Institute for Cancer Research, 1275 York Avenue, New York, NY 10065, USA; Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Lorenz Studer
- The Center for Stem Cell Biology, Sloan Kettering Institute for Cancer Research, 1275 York Avenue, New York, NY 10065, USA; Developmental Biology Program, Sloan Kettering Institute for Cancer Research, 1275 York Avenue, New York, NY 10065, USA; Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
22
|
Carreno G, Apps JR, Lodge EJ, Panousopoulos L, Haston S, Gonzalez-Meljem JM, Hahn H, Andoniadou CL, Martinez-Barbera JP. Hypothalamic sonic hedgehog is required for cell specification and proliferation of LHX3/LHX4 pituitary embryonic precursors. Development 2017; 144:3289-3302. [PMID: 28807898 DOI: 10.1242/dev.153387] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 08/01/2017] [Indexed: 12/31/2022]
Abstract
Sonic hedgehog (SHH) is an essential morphogenetic signal that dictates cell fate decisions in several developing organs in mammals. In vitro data suggest that SHH is required to specify LHX3+/LHX4+ Rathke's pouch (RP) progenitor identity. However, in vivo studies have failed to reveal such a function, supporting instead a crucial role for SHH in promoting proliferation of these RP progenitors and for differentiation of pituitary cell types. Here, we have used a genetic approach to demonstrate that activation of the SHH pathway is necessary to induce LHX3+/LHX4+ RP identity in mouse embryos. First, we show that conditional deletion of Shh in the anterior hypothalamus results in a fully penetrant phenotype characterised by a complete arrest of RP development, with lack of Lhx3/Lhx4 expression in RP epithelium at 9.0 days post coitum (dpc) and total loss of pituitary tissue by 12.5 dpc. Conversely, overactivation of the SHH pathway by conditional deletion of Ptch1 in RP progenitors leads to severe hyperplasia and enlargement of the Sox2+ stem cell compartment by the end of gestation.
Collapse
Affiliation(s)
- Gabriela Carreno
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - John R Apps
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Emily J Lodge
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Leonidas Panousopoulos
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Scott Haston
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Jose Mario Gonzalez-Meljem
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Heidi Hahn
- Institute of Human Genetics, Tumor Genetics Group, University of Göttingen, 37073 Göttingen, Germany
| | - Cynthia L Andoniadou
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK.,Department of Internal Medicine III, Technische Universität Dresden, 01307 Dresden, Germany
| | - Juan Pedro Martinez-Barbera
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| |
Collapse
|
23
|
Haston S, Pozzi S, Carreno G, Manshaei S, Panousopoulos L, Gonzalez-Meljem JM, Apps JR, Virasami A, Thavaraj S, Gutteridge A, Forshew T, Marais R, Brandner S, Jacques TS, Andoniadou CL, Martinez-Barbera JP. MAPK pathway control of stem cell proliferation and differentiation in the embryonic pituitary provides insights into the pathogenesis of papillary craniopharyngioma. Development 2017; 144:2141-2152. [PMID: 28506993 PMCID: PMC5482995 DOI: 10.1242/dev.150490] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 05/03/2017] [Indexed: 01/19/2023]
Abstract
Despite the importance of the RAS-RAF-MAPK pathway in normal physiology and disease of numerous organs, its role during pituitary development and tumourigenesis remains largely unknown. Here, we show that the over-activation of the MAPK pathway, through conditional expression of the gain-of-function alleles BrafV600E and KrasG12D in the developing mouse pituitary, results in severe hyperplasia and abnormal morphogenesis of the gland by the end of gestation. Cell-lineage commitment and terminal differentiation are disrupted, leading to a significant reduction in numbers of most of the hormone-producing cells before birth, with the exception of corticotrophs. Of note, Sox2+ stem cells and clonogenic potential are drastically increased in the mutant pituitaries. Finally, we reveal that papillary craniopharyngioma (PCP), a benign human pituitary tumour harbouring BRAF p.V600E also contains Sox2+ cells with sustained proliferative capacity and disrupted pituitary differentiation. Together, our data demonstrate a crucial function of the MAPK pathway in controlling the balance between proliferation and differentiation of Sox2+ cells and suggest that persistent proliferative capacity of Sox2+ cells may underlie the pathogenesis of PCP.
Collapse
Affiliation(s)
- Scott Haston
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Sara Pozzi
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Gabriela Carreno
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Saba Manshaei
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Leonidas Panousopoulos
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Jose Mario Gonzalez-Meljem
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - John R Apps
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Alex Virasami
- Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3NN, UK
| | - Selvam Thavaraj
- Head and Neck Pathology, Dental Institute, King's College London, London SE1 9RT, UK
| | - Alice Gutteridge
- Department of Pathology, UCL Cancer Institute, London WC1E 6DD, UK
| | - Tim Forshew
- Department of Pathology, UCL Cancer Institute, London WC1E 6DD, UK
| | - Richard Marais
- Molecular Oncology Group, Cancer Research UK Manchester Institute, The University of Manchester, Manchester M20 4BX, UK
| | - Sebastian Brandner
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London WC1N 3BG, UK
- Division of Neuropathology, The National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, UK
| | - Thomas S Jacques
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- Department of Histopathology, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3NN, UK
| | - Cynthia L Andoniadou
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
- Department of Internal Medicine III, Technische Universität Dresden, Dresden 01307, Germany
| | - Juan Pedro Martinez-Barbera
- Developmental Biology and Cancer Programme, Birth Defects Research Centre, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| |
Collapse
|
24
|
Zhu X, Tollkuhn J, Taylor H, Rosenfeld MG. Notch-Dependent Pituitary SOX2(+) Stem Cells Exhibit a Timed Functional Extinction in Regulation of the Postnatal Gland. Stem Cell Reports 2016; 5:1196-1209. [PMID: 26651607 PMCID: PMC4682291 DOI: 10.1016/j.stemcr.2015.11.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 11/02/2015] [Accepted: 11/02/2015] [Indexed: 01/17/2023] Open
Abstract
Although SOX2+ stem cells are present in the postnatal pituitary gland, how they are regulated molecularly and whether they are required for pituitary functions remain unresolved questions. Using a conditional knockout animal model, here we demonstrate that ablation of the canonical Notch signaling in the embryonic pituitary gland leads to progressive depletion of the SOX2+ stem cells and hypoplastic gland. Furthermore, we show that the SOX2+ stem cells initially play a significant role in contributing to postnatal pituitary gland expansion by self-renewal and differentiating into distinct lineages in the immediate postnatal period. However, we found that within several weeks postpartum, the SOX2+ stem cells switch to an essentially dormant state and are no longer required for homeostasis/tissue adaptation. Our results present a dynamic tissue homeostatic model in which stem cells provide an initial contribution to the growth of the neonatal pituitary gland, whereas the mature gland can be maintained in a stem cell-independent fashion. Notch signaling is necessary to maintain Sox2+ stem cells in the pituitary gland Sox2+ cells and differentiated cells contribute to postnatal pituitary expansion Sox2+ stem cells prove to be dispensable for adult pituitary gland homeostasis Differentiated cells retain mitotic capacity and respond to physiological demands
Collapse
Affiliation(s)
- Xiaoyan Zhu
- Howard Hughes Medical Institute, Department and School of Medicine, University of California at San Diego, La Jolla, CA 92093, USA.
| | - Jessica Tollkuhn
- Howard Hughes Medical Institute, Department and School of Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - Havilah Taylor
- Howard Hughes Medical Institute, Department and School of Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | - Michael G Rosenfeld
- Howard Hughes Medical Institute, Department and School of Medicine, University of California at San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
25
|
Stallings CE, Kapali J, Ellsworth BS. Mouse Models of Gonadotrope Development. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 143:1-48. [PMID: 27697200 DOI: 10.1016/bs.pmbts.2016.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The pituitary gonadotrope is central to reproductive function. Gonadotropes develop in a systematic process dependent on signaling factors secreted from surrounding tissues and those produced within the pituitary gland itself. These signaling pathways are important for stimulating specific transcription factors that ultimately regulate the expression of genes and define gonadotrope identity. Proper gonadotrope development and ultimately gonadotrope function are essential for normal sexual maturation and fertility. Understanding the mechanisms governing differentiation programs of gonadotropes is important to improve treatment and molecular diagnoses for patients with gonadotrope abnormalities. Much of what is known about gonadotrope development has been elucidated from mouse models in which important factors contributing to gonadotrope development and function have been deleted, ectopically expressed, or modified. This chapter will focus on many of these mouse models and their contribution to our current understanding of gonadotrope development.
Collapse
Affiliation(s)
- C E Stallings
- Department of Physiology, School of Medicine, Southern Illinois University, Carbondale, IL, United States
| | - J Kapali
- Department of Physiology, School of Medicine, Southern Illinois University, Carbondale, IL, United States
| | - B S Ellsworth
- Department of Physiology, School of Medicine, Southern Illinois University, Carbondale, IL, United States.
| |
Collapse
|
26
|
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
|
27
|
Aujla PK, Bogdanovic V, Naratadam GT, Raetzman LT. Persistent expression of activated notch in the developing hypothalamus affects survival of pituitary progenitors and alters pituitary structure. Dev Dyn 2016; 244:921-34. [PMID: 25907274 DOI: 10.1002/dvdy.24283] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 04/10/2015] [Accepted: 04/13/2015] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND As the pituitary gland develops, signals from the hypothalamus are necessary for pituitary induction and expansion. Little is known about the control of cues that regulate early signaling between the two structures. Ligands and receptors of the Notch signaling pathway are found in both the hypothalamus and Rathke's pouch. The downstream Notch effector gene Hes1 is required for proper pituitary formation; however, these effects could be due to the action of Hes1 in the hypothalamus, Rathke's pouch, or both. To determine the contribution of hypothalamic Notch signaling to pituitary organogenesis, we used mice with loss and gain of Notch function within the developing hypothalamus. RESULTS We demonstrate that loss of Notch signaling by conditional deletion of Rbpj in the hypothalamus does not affect expression of Hes1 within the posterior hypothalamus or expression of Hes5. In contrast, expression of activated Notch within the hypothalamus results in ectopic Hes5 expression and increased Hes1 expression, which is sufficient to disrupt pituitary development and postnatal expansion. CONCLUSIONS Taken together, our results indicate that Rbpj-dependent Notch signaling within the developing hypothalamus is not necessary for pituitary development, but persistent Notch signaling and ectopic Hes5 expression in hypothalamic progenitors affects pituitary induction and expansion.
Collapse
Affiliation(s)
- Paven K Aujla
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Vedran Bogdanovic
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - George T Naratadam
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Lori T Raetzman
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| |
Collapse
|
28
|
Yoshida S, Kato T, Kato Y. EMT Involved in Migration of Stem/Progenitor Cells for Pituitary Development and Regeneration. J Clin Med 2016; 5:jcm5040043. [PMID: 27058562 PMCID: PMC4850466 DOI: 10.3390/jcm5040043] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 03/24/2016] [Accepted: 03/29/2016] [Indexed: 12/17/2022] Open
Abstract
Epithelial–mesenchymal transition (EMT) and cell migration are important processes in embryonic development of many tissues as well as oncogenesis. The pituitary gland is a master endocrine tissue and recent studies indicate that Sox2-expressing stem/progenitor cells actively migrate and develop this tissue during embryogenesis. Notably, although migration activity of stem/progenitor cells in the postnatal period seems to be reduced compared to that in the embryonic period, it is hypothesized that stem/progenitor cells in the adult pituitary re-migrate from their microenvironment niche to contribute to the regeneration system. Therefore, elucidation of EMT in the pituitary stem/progenitor cells will promote understanding of pituitary development and regeneration, as well as diseases such as pituitary adenoma. In this review, so as to gain more insights into the mechanisms of pituitary development and regeneration, we summarize the EMT in the pituitary by focusing on the migration of pituitary stem/progenitor cells during both embryonic and postnatal organogenesis.
Collapse
Affiliation(s)
- Saishu Yoshida
- Organization for the Strategic Coordination of Research and Intellectual Property, Meiji University, Kanagawa 214-8571, Japan.
| | - Takako Kato
- Organization for the Strategic Coordination of Research and Intellectual Property, Meiji University, Kanagawa 214-8571, Japan.
- Institute of Reproduction and Endocrinology, Meiji University, Kanagawa 214-8571, Japan.
| | - Yukio Kato
- Institute of Reproduction and Endocrinology, Meiji University, Kanagawa 214-8571, Japan.
- Division of Life Science, Graduate School of Agriculture, Meiji University, Kanagawa 214-8571, Japan.
- Department of Life Science, School of Agriculture, Meiji University, Kanagawa 214-8571, Japan.
| |
Collapse
|
29
|
Davis SW, Keisler JL, Pérez-Millán MI, Schade V, Camper SA. All Hormone-Producing Cell Types of the Pituitary Intermediate and Anterior Lobes Derive From Prop1-Expressing Progenitors. Endocrinology 2016; 157:1385-96. [PMID: 26812162 PMCID: PMC4816735 DOI: 10.1210/en.2015-1862] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Mutations in PROP1, the most common known cause of combined pituitary hormone deficiency in humans, can result in the progressive loss of all hormones of the pituitary anterior lobe. In mice, Prop1 mutations result in the failure to initiate transcription of Pou1f1 (also known as Pit1) and lack somatotropins, lactotropins, and thyrotropins. The basis for this species difference is unknown. We hypothesized that Prop1 is expressed in a progenitor cell that can develop into all anterior lobe cell types, and not just the somatotropes, thyrotropes, and lactotropes, which are collectively known as the PIT1 lineage. To test this idea, we produced a transgenic Prop1-cre mouse line and conducted lineage-tracing experiments of Prop1-expressing cells. The results reveal that all hormone-secreting cell types of both the anterior and intermediate lobes are descended from Prop1-expressing progenitors. The Prop1-cre mice also provide a valuable genetic reagent with a unique spatial and temporal expression for generating tissue-specific gene rearrangements early in pituitary gland development. We also determined that the minimal essential sequences for reliable Prop1 expression lie within 10 kilobases of the mouse gene and demonstrated that human PROP1 can substitute functionally for mouse Prop1. These studies enhance our understanding of the pathophysiology of disease in patients with PROP1 mutations.
Collapse
Affiliation(s)
- Shannon W Davis
- Department of Biological Sciences (S.W.D., J.L.K.), University of South Carolina, Columbia, South Carolina 29208; and Department of Human Genetics (M.I.P.-M., V.S., S.A.C.), University of Michigan, Ann Arbor, Michigan 48109
| | - Jessica L Keisler
- Department of Biological Sciences (S.W.D., J.L.K.), University of South Carolina, Columbia, South Carolina 29208; and Department of Human Genetics (M.I.P.-M., V.S., S.A.C.), University of Michigan, Ann Arbor, Michigan 48109
| | - María I Pérez-Millán
- Department of Biological Sciences (S.W.D., J.L.K.), University of South Carolina, Columbia, South Carolina 29208; and Department of Human Genetics (M.I.P.-M., V.S., S.A.C.), University of Michigan, Ann Arbor, Michigan 48109
| | - Vanessa Schade
- Department of Biological Sciences (S.W.D., J.L.K.), University of South Carolina, Columbia, South Carolina 29208; and Department of Human Genetics (M.I.P.-M., V.S., S.A.C.), University of Michigan, Ann Arbor, Michigan 48109
| | - Sally A Camper
- Department of Biological Sciences (S.W.D., J.L.K.), University of South Carolina, Columbia, South Carolina 29208; and Department of Human Genetics (M.I.P.-M., V.S., S.A.C.), University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
30
|
Gaston-Massuet C, McCabe MJ, Scagliotti V, Young RM, Carreno G, Gregory LC, Jayakody SA, Pozzi S, Gualtieri A, Basu B, Koniordou M, Wu CI, Bancalari RE, Rahikkala E, Veijola R, Lopponen T, Graziola F, Turton J, Signore M, Mousavy Gharavy SN, Charolidi N, Sokol SY, Andoniadou CL, Wilson SW, Merrill BJ, Dattani MT, Martinez-Barbera JP. Transcription factor 7-like 1 is involved in hypothalamo-pituitary axis development in mice and humans. Proc Natl Acad Sci U S A 2016; 113:E548-57. [PMID: 26764381 PMCID: PMC4747739 DOI: 10.1073/pnas.1503346113] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Aberrant embryonic development of the hypothalamus and/or pituitary gland in humans results in congenital hypopituitarism (CH). Transcription factor 7-like 1 (TCF7L1), an important regulator of the WNT/β-catenin signaling pathway, is expressed in the developing forebrain and pituitary gland, but its role during hypothalamo-pituitary (HP) axis formation or involvement in human CH remains elusive. Using a conditional genetic approach in the mouse, we first demonstrate that TCF7L1 is required in the prospective hypothalamus to maintain normal expression of the hypothalamic signals involved in the induction and subsequent expansion of Rathke's pouch progenitors. Next, we reveal that the function of TCF7L1 during HP axis development depends exclusively on the repressing activity of TCF7L1 and does not require its interaction with β-catenin. Finally, we report the identification of two independent missense variants in human TCF7L1, p.R92P and p.R400Q, in a cohort of patients with forebrain and/or pituitary defects. We demonstrate that these variants exhibit reduced repressing activity in vitro and in vivo relative to wild-type TCF7L1. Together, our data provide support for a conserved molecular function of TCF7L1 as a transcriptional repressor during HP axis development in mammals and identify variants in this transcription factor that are likely to contribute to the etiology of CH.
Collapse
Affiliation(s)
- Carles Gaston-Massuet
- Birth Defects Research Centre, Developmental Biology and Cancer Programme, University College London Institute of Child Health, London, WC1N 1EH, United Kingdom
| | - Mark J McCabe
- Genetics and Epigenetics in Health and Disease Section, Genetics and Genomic Medicine Programme, University College London Institute of Child Health, London WC1N 1EH, United Kingdom
| | - Valeria Scagliotti
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Rodrigo M Young
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, United Kingdom
| | - Gabriela Carreno
- Birth Defects Research Centre, Developmental Biology and Cancer Programme, University College London Institute of Child Health, London, WC1N 1EH, United Kingdom
| | - Louise C Gregory
- Genetics and Epigenetics in Health and Disease Section, Genetics and Genomic Medicine Programme, University College London Institute of Child Health, London WC1N 1EH, United Kingdom
| | - Sujatha A Jayakody
- Birth Defects Research Centre, Developmental Biology and Cancer Programme, University College London Institute of Child Health, London, WC1N 1EH, United Kingdom
| | - Sara Pozzi
- Birth Defects Research Centre, Developmental Biology and Cancer Programme, University College London Institute of Child Health, London, WC1N 1EH, United Kingdom
| | - Angelica Gualtieri
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - Basudha Basu
- Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029
| | - Markela Koniordou
- Birth Defects Research Centre, Developmental Biology and Cancer Programme, University College London Institute of Child Health, London, WC1N 1EH, United Kingdom
| | - Chun-I Wu
- Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, Illinois, IL 60607
| | - Rodrigo E Bancalari
- Genetics and Epigenetics in Health and Disease Section, Genetics and Genomic Medicine Programme, University College London Institute of Child Health, London WC1N 1EH, United Kingdom
| | - Elisa Rahikkala
- Research Unit for Pediatrics, Dermatology, Clinical Genetics, Obstetrics and Gynecology (PEDEGO) and Medical Research Center (MRC) Oulu, University of Oulu, FIN-90029, Oulu, Finland; Department of Clinical Genetics, Oulu University Hospital, FIN-90029, Oulu, Finland
| | - Riitta Veijola
- Department of Pediatrics, PEDEGO and MRC Oulu, Oulu University Hospital, University of Oulu, FIN-90014, Oulu, Finland
| | - Tuija Lopponen
- Department of Child Neurology, Kuopio University Hospital, FIN 70029, Kuopio, Finland
| | - Federica Graziola
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, United Kingdom
| | - James Turton
- Genetics and Epigenetics in Health and Disease Section, Genetics and Genomic Medicine Programme, University College London Institute of Child Health, London WC1N 1EH, United Kingdom
| | - Massimo Signore
- Birth Defects Research Centre, Developmental Biology and Cancer Programme, University College London Institute of Child Health, London, WC1N 1EH, United Kingdom
| | - Seyedeh Neda Mousavy Gharavy
- Birth Defects Research Centre, Developmental Biology and Cancer Programme, University College London Institute of Child Health, London, WC1N 1EH, United Kingdom
| | - Nicoletta Charolidi
- Birth Defects Research Centre, Developmental Biology and Cancer Programme, University College London Institute of Child Health, London, WC1N 1EH, United Kingdom
| | - Sergei Y Sokol
- Developmental and Regenerative Biology, Mount Sinai School of Medicine, New York, NY 10029
| | - Cynthia Lilian Andoniadou
- Birth Defects Research Centre, Developmental Biology and Cancer Programme, University College London Institute of Child Health, London, WC1N 1EH, United Kingdom
| | - Stephen W Wilson
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, United Kingdom
| | - Bradley J Merrill
- Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, Illinois, IL 60607
| | - Mehul T Dattani
- Genetics and Epigenetics in Health and Disease Section, Genetics and Genomic Medicine Programme, University College London Institute of Child Health, London WC1N 1EH, United Kingdom
| | - Juan Pedro Martinez-Barbera
- Birth Defects Research Centre, Developmental Biology and Cancer Programme, University College London Institute of Child Health, London, WC1N 1EH, United Kingdom;
| |
Collapse
|
31
|
Affiliation(s)
- Buffy S Ellsworth
- Department of Physiology, Southern Illinois University, Carbondale, Illinois 62901-6523
| |
Collapse
|
32
|
Schoenmakers N, Alatzoglou KS, Chatterjee VK, Dattani MT. Recent advances in central congenital hypothyroidism. J Endocrinol 2015; 227:R51-71. [PMID: 26416826 PMCID: PMC4629398 DOI: 10.1530/joe-15-0341] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Revised: 09/17/2015] [Accepted: 09/28/2015] [Indexed: 01/23/2023]
Abstract
Central congenital hypothyroidism (CCH) may occur in isolation, or more frequently in combination with additional pituitary hormone deficits with or without associated extrapituitary abnormalities. Although uncommon, it may be more prevalent than previously thought, affecting up to 1:16 000 neonates in the Netherlands. Since TSH is not elevated, CCH will evade diagnosis in primary, TSH-based, CH screening programs and delayed detection may result in neurodevelopmental delay due to untreated neonatal hypothyroidism. Alternatively, coexisting growth hormones or ACTH deficiency may pose additional risks, such as life threatening hypoglycaemia. Genetic ascertainment is possible in a minority of cases and reveals mutations in genes controlling the TSH biosynthetic pathway (TSHB, TRHR, IGSF1) in isolated TSH deficiency, or early (HESX1, LHX3, LHX4, SOX3, OTX2) or late (PROP1, POU1F1) pituitary transcription factors in combined hormone deficits. Since TSH cannot be used as an indicator of euthyroidism, adequacy of treatment can be difficult to monitor due to a paucity of alternative biomarkers. This review will summarize the normal physiology of pituitary development and the hypothalamic-pituitary-thyroid axis, then describe known genetic causes of isolated central hypothyroidism and combined pituitary hormone deficits associated with TSH deficiency. Difficulties in diagnosis and management of these conditions will then be discussed.
Collapse
Affiliation(s)
- Nadia Schoenmakers
- University of Cambridge Metabolic Research LaboratoriesWellcome Trust-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Level 4, PO Box 289, Hills Road, Cambridge CB2 0QQ, UKDevelopmental Endocrinology Research GroupSection of Genetics and Epigenetics in Health and Disease, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London, UK
| | - Kyriaki S Alatzoglou
- University of Cambridge Metabolic Research LaboratoriesWellcome Trust-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Level 4, PO Box 289, Hills Road, Cambridge CB2 0QQ, UKDevelopmental Endocrinology Research GroupSection of Genetics and Epigenetics in Health and Disease, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London, UK
| | - V Krishna Chatterjee
- University of Cambridge Metabolic Research LaboratoriesWellcome Trust-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Level 4, PO Box 289, Hills Road, Cambridge CB2 0QQ, UKDevelopmental Endocrinology Research GroupSection of Genetics and Epigenetics in Health and Disease, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London, UK
| | - Mehul T Dattani
- University of Cambridge Metabolic Research LaboratoriesWellcome Trust-Medical Research Council Institute of Metabolic Science, Addenbrooke's Hospital, Level 4, PO Box 289, Hills Road, Cambridge CB2 0QQ, UKDevelopmental Endocrinology Research GroupSection of Genetics and Epigenetics in Health and Disease, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London, UK
| |
Collapse
|
33
|
Ochiai H, Suga H, Yamada T, Sakakibara M, Kasai T, Ozone C, Ogawa K, Goto M, Banno R, Tsunekawa S, Sugimura Y, Arima H, Oiso Y. BMP4 and FGF strongly induce differentiation of mouse ES cells into oral ectoderm. Stem Cell Res 2015; 15:290-8. [PMID: 26209816 DOI: 10.1016/j.scr.2015.06.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 06/08/2015] [Accepted: 06/30/2015] [Indexed: 11/30/2022] Open
Abstract
During embryonic development, oral ectoderm differentiates into the adenohypophysis, dental epithelia, salivary glands, and nasal pit. Few reports exist concerning the induction of oral ectoderm from embryonic stem (ES) cells. Generally, any lot differences in fetal bovine serum (FBS) and serum replacer may affect the induction of ES cell-differentiation. Using a previously established culture strategy for differentiation, the proportion of cell aggregates containing Pitx1+ oral ectoderm varied widely between 9-36% when several different lots of FBS or serum replacer were used. We therefore tried to enhance the differentiation method. We found that bone morphogenetic protein (BMP) 4 and fibroblast growth factor (FGF) treatments improved oral ectoderm induction. Such treatment also improved the differentiation of oral ectoderm into the adenohypophysis. Furthermore, increased BMP4 treatment induced dental epithelium and mesenchyme. Such differentiation suggests that the Pitx1+ layer displays similar properties to oral ectoderm, as found in vivo. Differentiation of ES cells into oral ectoderm using different lots of FBS and serum replacer increased 78-90% after treatment with BMP4 and FGF. In summary, we have established a robust strategy for the induction of oral ectoderm differentiation from mouse ES cells.
Collapse
Affiliation(s)
- Hiroshi Ochiai
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan.
| | - Tomiko Yamada
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Mayu Sakakibara
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Takatoshi Kasai
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Chikafumi Ozone
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Koichiro Ogawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Motomitsu Goto
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Ryoichi Banno
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Shin Tsunekawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yoshihisa Sugimura
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Hiroshi Arima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yutaka Oiso
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsuruma-cho, Showa-ku, Nagoya 466-8550, Japan
| |
Collapse
|
34
|
Martinez-Barbera JP. 60 YEARS OF NEUROENDOCRINOLOGY: Biology of human craniopharyngioma: lessons from mouse models. J Endocrinol 2015; 226:T161-72. [PMID: 25926515 DOI: 10.1530/joe-15-0145] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/29/2015] [Indexed: 01/29/2023]
Abstract
Adamantinomatous craniopharyngiomas (ACP) are clinically relevant tumours that are associated with high morbidity, poor quality of life and occasional mortality. Human and mouse studies have provided important insights into the biology of these aggressive tumours, and we are starting to understand why, how and when these tumours develop in humans. Mutations in β-catenin that result in the over-activation of the WNT/β-catenin signalling pathway are critical drivers of most, perhaps of all, human ACPs. Mouse studies have shown that only pituitary embryonic precursors or adult stem cells are able to generate tumours when targeted with oncogenic β-catenin, which suggests that the cell context is critical in order for mutant β-catenin to exert its oncogenic effect. Interestingly, mutant stem cells do not generate the bulk of the tumour cells; instead, they induce tumours in a paracrine manner. Combining basic studies in mice and humans will provide further insights into the biology of these neoplasms and will reveal pathogenic pathways that could be targeted with specific inhibitors for the benefit of patients. These benign tumours may additionally represent a unique model for investigating the early steps that lead to oncogenesis.
Collapse
Affiliation(s)
- Juan Pedro Martinez-Barbera
- Developmental Biology and Cancer ProgrammeInstitute of Child Health, Birth Defects Research Centre, University College London, 30 Guilford Street, WC1N 1EH London, UK
| |
Collapse
|
35
|
Musumeci G, Castorina S, Castrogiovanni P, Loreto C, Leonardi R, Aiello FC, Magro G, Imbesi R. A journey through the pituitary gland: Development, structure and function, with emphasis on embryo-foetal and later development. Acta Histochem 2015; 117:355-66. [PMID: 25858531 DOI: 10.1016/j.acthis.2015.02.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 12/28/2014] [Accepted: 02/05/2015] [Indexed: 12/22/2022]
Abstract
The pituitary gland and the hypothalamus are morphologically and functionally associated in the endocrine and neuroendocrine control of other endocrine glands. They therefore play a key role in a number of regulatory feedback processes that co-ordinate the whole endocrine system. Here we review the neuroendocrine system, from the discoveries that led to its identification to some recently clarified embryological, functional, and morphological aspects. In particular we review the pituitary gland and the main notions related to its development, organization, cell differentiation, and vascularization. Given the crucial importance of the factors controlling neuroendocrine system development to understand parvocellular neuron function and the aetiology of the congenital disorders related to hypothalamic-pituitary axis dysfunction, we also provide an overview of the molecular and genetic studies that have advanced our knowledge in the field. Through the action of the hypothalamus, the pituitary gland is involved in the control of a broad range of key aspects of our lives: the review focuses on the hypothalamic-pituitary-gonadal axis, particularly GnRH, whose abnormal secretion is associated with clinical conditions involving delayed or absent puberty and reproductive dysfunction.
Collapse
|
36
|
Abstract
Significant progress has been made recently in unravelling the embryonic events leading to pituitary morphogenesis, both in vivo and in vitro. This includes dissection of the molecular mechanisms controlling patterning of the ventral diencephalon that regulate formation of the pituitary anlagen or Rathke's pouch. There is also a better characterisation of processes that underlie maintenance of pituitary progenitors, specification of endocrine lineages and the three-dimensional organisation of newly differentiated endocrine cells. Furthermore, a population of adult pituitary stem cells (SCs), originating from embryonic progenitors, have been described and shown to have not only regenerative potential, but also the capacity to induce tumour formation. Finally, the successful recapitulation in vitro of embryonic events leading to generation of endocrine cells from embryonic SCs, and their subsequent transplantation, represents exciting advances towards the use of regenerative medicine to treat endocrine deficits. In this review, an up-to-date description of pituitary morphogenesis will be provided and discussed with particular reference to pituitary SC studies.
Collapse
Affiliation(s)
- Karine Rizzoti
- Division of Stem Cell Biology and Developmental GeneticsMRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK
| |
Collapse
|
37
|
Alatzoglou KS, Webb EA, Le Tissier P, Dattani MT. Isolated growth hormone deficiency (GHD) in childhood and adolescence: recent advances. Endocr Rev 2014; 35:376-432. [PMID: 24450934 DOI: 10.1210/er.2013-1067] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The diagnosis of GH deficiency (GHD) in childhood is a multistep process involving clinical history, examination with detailed auxology, biochemical testing, and pituitary imaging, with an increasing contribution from genetics in patients with congenital GHD. Our increasing understanding of the factors involved in the development of somatotropes and the dynamic function of the somatotrope network may explain, at least in part, the development and progression of childhood GHD in different age groups. With respect to the genetic etiology of isolated GHD (IGHD), mutations in known genes such as those encoding GH (GH1), GHRH receptor (GHRHR), or transcription factors involved in pituitary development, are identified in a relatively small percentage of patients suggesting the involvement of other, yet unidentified, factors. Genome-wide association studies point toward an increasing number of genes involved in the control of growth, but their role in the etiology of IGHD remains unknown. Despite the many years of research in the area of GHD, there are still controversies on the etiology, diagnosis, and management of IGHD in children. Recent data suggest that childhood IGHD may have a wider impact on the health and neurodevelopment of children, but it is yet unknown to what extent treatment with recombinant human GH can reverse this effect. Finally, the safety of recombinant human GH is currently the subject of much debate and research, and it is clear that long-term controlled studies are needed to clarify the consequences of childhood IGHD and the long-term safety of its treatment.
Collapse
Affiliation(s)
- Kyriaki S Alatzoglou
- Developmental Endocrinology Research Group (K.S.A., E.A.W., M.T.D.), Clinical and Molecular Genetics Unit, and Birth Defects Research Centre (P.L.T.), UCL Institute of Child Health, London WC1N 1EH, United Kingdom; and Faculty of Life Sciences (P.L.T.), University of Manchester, Manchester M13 9PT, United Kingdom
| | | | | | | |
Collapse
|
38
|
Effects of diethylstilbestrol on luteinizing hormone-producing cells in the mouse anterior pituitary. Exp Biol Med (Maywood) 2014; 239:311-9. [DOI: 10.1177/1535370213519722] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Gonadotrophs in the anterior pituitary secrete luteinizing hormone (LH) and follicle-stimulating hormone (FSH). Neonatal diethylstilbestrol (neoDES) treatment affects reproductive function of male and female mice, but the effect of this treatment on the development as well as direct effects on pituitary gonadotrophs have not been ascertained. We investigated LH-secreting gonadotropes and the expression of genes involved in the synthesis and secretion of gonadotropins in the anterior pituitary of neoDES mice using immunohistochemistry and real-time reverse transcription-polymerase chain reaction (RT-PCR). The percentage of LH-secreting gonadotropes in 90-day-old female mice treated neonatally with an oil vehicle (neoOil) was significantly lower than in 30-day-old neoOil females but not in males, indicating a significant reduction after reproductive maturation in females. The percentage of LH-secreting gonadotropes in the medial area of 90-day-old neoDES females was significantly lower than that of 90-day-old neoOil females, ovariectomized neoOil females, and neoOil and neoDES males. The expression of the LH beta ( Lhb) subunit in the anterior pituitary of 90-day-old neoDES females was similar to that in neoOil females, but it was significantly lower than that observed in 90-day-old males. Ovariectomy increased the expression of the alpha subunit of glycoprotein hormones, FSH beta ( Fshb) subunit and Lhb subunit both in neoOil and neoDES females, suggesting that the anterior pituitary of neoDES female mice is regulated by ovarian hormones via negative feedback. In organ-cultured, anterior pituitaries exposed to DES exhibited no change in the number of LH-secreting gonadotropes but did reduced gene expression. These results suggest that LH-secreting gonadotropes in the female mice are not only directly affected by neoDES but also are influenced by the masculinization of the hypothalamus. That is, neonatal DES exposure can masculinize or defeminize the hypothalamus of female mice. However, the regulation of the pituitary gonadotropins by the hypothalamus could be different from that in intact male mice.
Collapse
|
39
|
Andoniadou CL, Matsushima D, Mousavy Gharavy SN, Signore M, Mackintosh AI, Schaeffer M, Gaston-Massuet C, Mollard P, Jacques TS, Le Tissier P, Dattani MT, Pevny LH, Martinez-Barbera JP. Sox2(+) stem/progenitor cells in the adult mouse pituitary support organ homeostasis and have tumor-inducing potential. Cell Stem Cell 2013; 13:433-45. [PMID: 24094324 DOI: 10.1016/j.stem.2013.07.004] [Citation(s) in RCA: 202] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 06/03/2013] [Accepted: 07/03/2013] [Indexed: 01/16/2023]
Abstract
Sox2(+) adult mouse pituitary cells can self-renew and terminally differentiate in vitro, but their physiological role in vivo and possible contribution to oncogenesis remain largely unknown. Using genetic lineage tracing, we show here that the Sox2(+) cell compartment of both the embryonic and adult pituitary contains stem/progenitor cells that are able to differentiate into all hormone-producing lineages and contribute to organ homeostasis during postnatal life. In addition, we show that targeted expression of oncogenic β-catenin in Sox2(+) cells gives rise to pituitary tumors, but, unexpectedly, the tumor mass is not derived from the Sox2(+) mutation-sustaining cells, suggesting a paracrine role of Sox2(+) cells in pituitary oncogenesis. Our data therefore provide in vivo evidence of a role for Sox2(+) stem/progenitor cells in long-term physiological maintenance of the adult pituitary, and highlight an unexpected non-cell-autonomous role for these cells in the induction of pituitary tumors.
Collapse
Affiliation(s)
- Cynthia Lilian Andoniadou
- Birth Defects Research Centre, Neural Development Unit, UCL Institute of Child Health, London, WC1N 1EH, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Rizzoti K, Akiyama H, Lovell-Badge R. Mobilized adult pituitary stem cells contribute to endocrine regeneration in response to physiological demand. Cell Stem Cell 2013; 13:419-32. [PMID: 24094323 PMCID: PMC3793864 DOI: 10.1016/j.stem.2013.07.006] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 06/07/2013] [Accepted: 07/14/2013] [Indexed: 12/18/2022]
Abstract
Pituitary hormone deficiencies, with Growth Hormone deficiency being most frequent (1 in 3,500-10,000 births), cause significant morbidity. Regeneration of missing endocrine cells would be a significant improvement over hormone replacement therapies, which incur side effects and do not mimic physiological secretion patterns. Recent in vitro studies have identified a population of adult pituitary progenitors that express the HMG box transcription factors SOX2 and SOX9. Here, we apply cell-lineage tracing analysis to demonstrate that SOX2- and SOX9-expressing progenitors can self-renew and give rise to endocrine cells in vivo, suggesting that they are tissue stem cells. Moreover, we show that they can become mobilized and differentiate into the appropriate endocrine cell types in response to physiological stress. Our results highlight the pituitary as a model for exploring how physiological changes influence stem cell behavior and suggest that manipulation of endogenous pituitary stem cells is a potential therapeutic strategy for pituitary deficiencies.
Collapse
Affiliation(s)
- Karine Rizzoti
- Division of Stem Cell Biology and Developmental Genetics, MRC National Institute for Medical Research, London NW7 1AA, UK.
| | | | | |
Collapse
|
41
|
Chen M, Kato T, Higuchi M, Yoshida S, Yako H, Kanno N, Kato Y. Coxsackievirus and adenovirus receptor-positive cells compose the putative stem/progenitor cell niches in the marginal cell layer and parenchyma of the rat anterior pituitary. Cell Tissue Res 2013; 354:823-36. [DOI: 10.1007/s00441-013-1713-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 07/16/2013] [Indexed: 01/04/2023]
|
42
|
Pérez-Millán MI, Zeidler MG, Saunders TL, Camper SA, Davis SW. Efficient, specific, developmentally appropriate cre-mediated recombination in anterior pituitary gonadotropes and thyrotropes. Genesis 2013; 51:785-92. [PMID: 23996951 DOI: 10.1002/dvg.22425] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 07/30/2013] [Accepted: 08/05/2013] [Indexed: 11/06/2022]
Abstract
Tissue-specific expression of cre recombinase is a well-established genetic tool to analyze gene function, and it is limited only by the efficiency and specificity of available cre mouse strains. Here, we report the generation of a transgenic line containing a cre cassette with codon usage optimized for mammalian cells (iCre) under the control of a mouse glycoprotein hormone α-subunit (αGSU) regulatory sequences in a bacterial artificial chromosome genomic clone. Initial analysis of this transgenic line, Tg(αGSU-iCre), with cre reporter strains reveals onset of cre activity in the differentiating cells of the developing anterior pituitary gland at embryonic day 12.5, with a pattern characteristic of endogenous αGSU. In adult mice, αGSU-iCre was active in the anterior lobe of the pituitary gland and in the cells that produce αGSU (gonadotropes and thyrotropes) with high penetrance. Little or no activity was observed in other tissues, including skeletal and cardiac muscle, brain, kidney, lungs, testis, ovary, and liver. This αGSU-iCre line is suitable for efficient, specific, and developmentally regulated deletion of floxed alleles in anterior pituitary gonadotropes and thyrotropes.
Collapse
|
43
|
Langlais D, Couture C, Kmita M, Drouin J. Adult pituitary cell maintenance: lineage-specific contribution of self-duplication. Mol Endocrinol 2013; 27:1103-12. [PMID: 23754801 PMCID: PMC4486415 DOI: 10.1210/me.2012-1407] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 05/27/2013] [Indexed: 11/19/2022] Open
Abstract
The identification of a stable pool of progenitor/stem cells in the adult pituitary has renewed the interest of identifying mechanisms for maintenance of pituitary cells throughout life. Whereas developmental studies have shown that progenitor expansion is the major source of new differentiated cells during pituitary organogenesis, the contribution of these progenitors for maintenance of the adult tissue is not clear although progenitors were clearly involved in cell expansion following end-organ ablation, notably after adrenalectomy and/or gonadectomy. We have used a genetic trick that eliminates dividing cells by apoptosis in order to assess the contribution of differentiated corticotropes and melanotropes for maintenance of their population in the adult pituitary. The system relies on chromosome instability created by the action of the Cre recombinase on inverted loxP sites. Expression of Cre recombinase in corticotropes and melanotropes led to progressive loss of corticotropes whereas melanotropes were unaffected. Because the Cre transgene is not expressed in progenitors, the data indicate that maintenance of the adult corticotrope pool is primarily due to self-duplication of differentiated cells. In contrast, melanotropes do not divide. Maintenance of corticotropes by self-duplication contrasts with the reported proliferative response of undifferentiated cells observed after adrenalectomy. If corticotrope reentry into cell cycle constitutes a normal mechanism to maintain the adult corticotrope pool, this same mechanism may also be perturbed during corticotrope adenoma development in Cushing's disease.
Collapse
Affiliation(s)
- David Langlais
- Laboratoire de génétique moléculaire, Institut de recherches cliniques de Montréal (IRCM), 110, avenue des Pins Ouest, Montréal, Quebec, H2W 1R7 Canada
| | | | | | | |
Collapse
|
44
|
Sasai Y, Eiraku M, Suga H. In vitro organogenesis in three dimensions: self-organising stem cells. Development 2013; 139:4111-21. [PMID: 23093423 DOI: 10.1242/dev.079590] [Citation(s) in RCA: 134] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Organ formation during embryogenesis is a complex process that involves various local cell-cell interactions at the molecular and mechanical levels. Despite this complexity, organogenesis can be modelled in vitro. In this article, we focus on two recent examples in which embryonic stem cells can self-organise into three-dimensional structures - the optic cup and the pituitary epithelium; and one case of self-organising adult stem cells - the gut epithelium. We summarise how these approaches have revealed intrinsic programs that drive locally autonomous modes of organogenesis and homeostasis. We also attempt to interpret the results of previous in vivo studies of retinal development in light of the self-organising nature of the retina.
Collapse
Affiliation(s)
- Yoshiki Sasai
- Neurogenesis and Organogenesis Group, RIKEN Center for Developmental Biology, Kobe, Japan.
| | | | | |
Collapse
|
45
|
Davis SW, Ellsworth BS, Peréz Millan MI, Gergics P, Schade V, Foyouzi N, Brinkmeier ML, Mortensen AH, Camper SA. Pituitary gland development and disease: from stem cell to hormone production. Curr Top Dev Biol 2013; 106:1-47. [PMID: 24290346 DOI: 10.1016/b978-0-12-416021-7.00001-8] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Many aspects of pituitary development have become better understood in the past two decades. The signaling pathways regulating pituitary growth and shape have emerged, and the balancing interactions between the pathways are now appreciated. Markers for multipotent progenitor cells are being identified, and signature transcription factors have been discovered for most hormone-producing cell types. We now realize that pulsatile hormone secretion involves a 3D integration of cellular networks. About a dozen genes are known to cause pituitary hypoplasia when mutated due to their essential roles in pituitary development. Similarly, a few genes are known that predispose to familial endocrine neoplasia, and several genes mutated in sporadic pituitary adenomas are documented. In the next decade, we anticipate gleaning a deeper appreciation of these processes at the molecular level, insight into the development of the hypophyseal portal blood system, and evolution of better therapeutics for congenital and acquired hormone deficiencies and for common craniopharyngiomas and pituitary adenomas.
Collapse
Affiliation(s)
- Shannon W Davis
- Department of Biological Sciences, University of South Carolina, Columbia, South Carolina, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Bouchoucha YX, Charnay P, Gilardi-Hebenstreit P. Ablation of Egr2-positive cells in male mouse anterior pituitary leads to atypical isolated GH deficiency. Endocrinology 2013; 154:270-82. [PMID: 23150495 DOI: 10.1210/en.2012-1792] [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] [Indexed: 11/19/2022]
Abstract
In this study, we have investigated the expression and function of the transcription factor early growth response factor 2 (Egr2)/Krox20 in the developing anterior pituitary. Egr2 is initially expressed in all differentiating hormonal cells types, but its expression is mostly restricted to the somatotroph lineage after birth. Egr2 knockout results in anterior pituitary hypoplasia. However, the analysis of a conditional mutant demonstrates that this phenotype does not originate from a lack of Egr2 expression in the pituitary. Using an Egr2 allele driving a Cre-activable toxin gene, we performed a genetic ablation of Egr2-positive cells in the pituitary. During the postnatal period, this ablation leads to specific and progressive depletion of the somatotroph population, creating a novel model of early-onset isolated GH deficiency (GHD). Mutant animals were subjected to a complete metabolic analysis, revealing atypical and expected features. Consistent with an adult-onset isolated GHD model, mutant animals are hypoglycemic and display increased insulin sensitivity and glucose clearance. This latter phenotype is in contrast to the glucose intolerance observed in another early-onset GHD model. Surprisingly, increased insulin sensitivity is not accompanied by a modified balance between fat and lean tissues, but by reduced metabolic adaptability between glucose and lipid oxidation conditions. This suggests that the relationship between these metabolic features and insulin sensitivity should be reconsidered. In conclusion, our mutant may be a valuable genetic model with which to study the effects of long-term GH deficiency, in conditions of normal pancreatic function and unaffected balance between fat and glucose metabolism.
Collapse
|
47
|
Majumdar S, Farris CL, Kabat BE, Jung DO, Ellsworth BS. Forkhead Box O1 is present in quiescent pituitary cells during development and is increased in the absence of p27 Kip1. PLoS One 2012; 7:e52136. [PMID: 23251696 PMCID: PMC3522653 DOI: 10.1371/journal.pone.0052136] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 11/15/2012] [Indexed: 02/07/2023] Open
Abstract
Congenital pituitary hormone deficiencies have been reported in approximately one in 4,000 live births, however studies reporting mutations in some widely studied transcription factors account for only a fraction of congenital hormone deficiencies in humans. Anterior pituitary hormones are required for development and function of several glands including gonads, adrenals, and thyroid. In order to identify additional factors that contribute to human congenital hormone deficiencies, we are investigating the forkhead transcription factor, FOXO1, which has been implicated in development of several organs including ovary, testis, and brain. We find that FOXO1 is present in the nuclei of non-dividing pituitary cells during embryonic development, consistent with a role in limiting proliferation and/or promoting differentiation. FOXO1 is present in a subset of differentiated cells at e18.5 and in adult with highest level of expression in somatotrope cells. We detected FOXO1 in p27(Kip1)-positive cells at e14.5. In the absence of p27(Kip1) the number of pituitary cells containing FOXO1 is significantly increased at e14.5 suggesting that a feedback loop regulates the interplay between FOXO1 and p27(Kip1).
Collapse
Affiliation(s)
- Sreeparna Majumdar
- Department of Physiology, Southern Illinois University, Carbondale, Illinois, United States of America
| | - Corrie L. Farris
- Department of Physiology, Southern Illinois University, Carbondale, Illinois, United States of America
| | - Brock E. Kabat
- Department of Physiology, Southern Illinois University, Carbondale, Illinois, United States of America
| | - Deborah O. Jung
- Department of Physiology, Southern Illinois University, Carbondale, Illinois, United States of America
| | - Buffy S. Ellsworth
- Department of Physiology, Southern Illinois University, Carbondale, Illinois, United States of America
| |
Collapse
|
48
|
Jayakody SA, Andoniadou CL, Gaston-Massuet C, Signore M, Cariboni A, Bouloux PM, Le Tissier P, Pevny LH, Dattani MT, Martinez-Barbera JP. SOX2 regulates the hypothalamic-pituitary axis at multiple levels. J Clin Invest 2012; 122:3635-46. [PMID: 22945632 DOI: 10.1172/jci64311] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 07/17/2012] [Indexed: 12/30/2022] Open
Abstract
Sex-determining region Y (SRY) box 2 (SOX2) haploinsufficiency causes a form of hypopituitarism in humans that is characterized by gonadotrophin deficiency known as hypogonadotrophic hypogonadism. Here, we conditionally deleted Sox2 in mice to investigate the pathogenesis of hypogonadotrophic hypogonadism. First, we found that absence of SOX2 in the developing Rathke pouch of conditional embryos led to severe anterior lobe hypoplasia with drastically reduced expression of the pituitary-specific transcription factor POU class 1 homeobox 1 (POU1F1) as well as severe disruption of somatotroph and thyrotroph differentiation. In contrast, corticotrophs, rostral-tip POU1F1-independent thyrotrophs, and, interestingly, lactotrophs and gonadotrophs were less affected. Second, we identified a requirement for SOX2 in normal proliferation of periluminal progenitors; in its absence, insufficient precursors were available to produce all cell lineages of the anterior pituitary. Differentiated cells derived from precursors exiting cell cycle at early stages, including corticotrophs, rostral-tip thyrotrophs, and gonadotrophs, were generated, while hormone-producing cells originating from late-born precursors, such as somatotrophs and POU1F1-dependent thyrotrophs, were severely reduced. Finally, we found that 2 previously characterized patients with SOX2 haploinsufficiency and associated hypogonadotrophic hypogonadism had a measurable response to gonadotropin-releasing hormone (GnRH) stimulation, suggesting that it is not the absence of gonadotroph differentiation, but rather the deficient hypothalamic stimulation of gonadotrophs, that underlies typical hypogonadotrophic hypogonadism.
Collapse
Affiliation(s)
- Sujatha A Jayakody
- Neural Development Unit, Institute of Child Health, and Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Le Tissier PR, Hodson DJ, Lafont C, Fontanaud P, Schaeffer M, Mollard P. Anterior pituitary cell networks. Front Neuroendocrinol 2012; 33:252-66. [PMID: 22981652 DOI: 10.1016/j.yfrne.2012.08.002] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 08/17/2012] [Accepted: 08/18/2012] [Indexed: 12/17/2022]
Abstract
Both endocrine and non-endocrine cells of the pituitary gland are organized into structural and functional networks which are formed during embryonic development but which may be modified throughout life. Structural mapping of the various endocrine cell types has highlighted the existence of distinct network motifs and relationships with the vasculature which may relate to temporal differences in their output. Functional characterization of the network activity of growth hormone and prolactin cells has revealed a role for cell organization in gene regulation, the plasticity of pituitary hormone output and remarkably the ability to memorize altered demand. As such, the description of these endocrine cell networks alters the concept of the pituitary from a gland which simply responds to external regulation to that of an oscillator which may memorize information and constantly adapt its coordinated networks' responses to the flow of hypothalamic inputs.
Collapse
Affiliation(s)
- P R Le Tissier
- Division of Molecular Neuroendocrinology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, United Kingdom;
| | | | | | | | | | | |
Collapse
|
50
|
Mollard P, Hodson DJ, Lafont C, Rizzoti K, Drouin J. A tridimensional view of pituitary development and function. Trends Endocrinol Metab 2012; 23:261-9. [PMID: 22436593 DOI: 10.1016/j.tem.2012.02.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Revised: 02/10/2012] [Accepted: 02/13/2012] [Indexed: 01/05/2023]
Abstract
Recent advances in tridimensional (3D) tissue imaging have considerably enriched our view of the pituitary gland and its development. Whereas traditional histology of the pituitary anterior lobe portrayed this tissue as a patchwork of cells, 3D imaging revealed that cells of each lineage form extensive and structured homotypic networks. In the adult gland these networks contribute to the robustness and coordination of the cell response to secretagogs. In addition, the network organization adapts to changes in endocrine environment, as revealed by the sexually dimorphic growth hormone (GH) cell network. Further work is required to establish better the molecular basis for homotypic and heterotypic interactions in the pituitary as well as the implications of these interactions for pituitary function and dysfunction in humans.
Collapse
Affiliation(s)
- Patrice Mollard
- Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 5203, Institut de Génomique Fonctionnelle, 34000 Montpellier, France.
| | | | | | | | | |
Collapse
|