1
|
Khetchoumian K, Sochodolsky K, Lafont C, Gouhier A, Bemmo A, Kherdjemil Y, Kmita M, Le Tissier P, Mollard P, Christian H, Drouin J. Paracrine FGF1 signaling directs pituitary architecture and size. Proc Natl Acad Sci U S A 2024; 121:e2410269121. [PMID: 39320918 PMCID: PMC11459159 DOI: 10.1073/pnas.2410269121] [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: 05/22/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024] Open
Abstract
Organ architecture is established during development through intricate cell-cell communication mechanisms, yet the specific signals mediating these communications often remain elusive. Here, we used the anterior pituitary gland that harbors different interdigitated hormone-secreting homotypic cell networks to dissect cell-cell communication mechanisms operating during late development. We show that blocking differentiation of corticotrope cells leads to pituitary hypoplasia with a major effect on somatotrope cells that directly contact corticotropes. Gene knockout of the corticotrope-restricted transcription factor Tpit results in fewer somatotropes, with less secretory granules and a loss of cell polarity, resulting in systemic growth retardation. Single-cell transcriptomic analyses identified FGF1 as a corticotrope-specific Tpit dosage-dependent target gene responsible for these phenotypes. Consistently, genetic ablation of FGF1 in mice phenocopies pituitary hypoplasia and growth impairment observed in Tpit-deficient mice. These findings reveal FGF1 produced by the corticotrope cell network as an essential paracrine signaling molecule participating in pituitary architecture and size.
Collapse
Affiliation(s)
- Konstantin Khetchoumian
- Laboratoire de génétique moléculaire, Institut de recherches cliniques de Montréal, Montréal, QCH2W 1R7, Canada
| | - Kevin Sochodolsky
- Laboratoire de génétique moléculaire, Institut de recherches cliniques de Montréal, Montréal, QCH2W 1R7, Canada
| | - Chrystel Lafont
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, MontpellierF-34094, France
- BioCampus Montpellier, University of Montpellier, CNRS, INSERM, MontpellierF-34094, France
| | - Arthur Gouhier
- Laboratoire de génétique moléculaire, Institut de recherches cliniques de Montréal, Montréal, QCH2W 1R7, Canada
| | - Amandine Bemmo
- Laboratoire de génétique moléculaire, Institut de recherches cliniques de Montréal, Montréal, QCH2W 1R7, Canada
| | - Yacine Kherdjemil
- Disease Modeling and Genome Editing platform, Institut de recherches cliniques de Montréal, Montréal, QCH2W 1R7, Canada
| | - Marie Kmita
- Laboratoire de recherche en génétique et développement, Institut de recherches cliniques de Montréal, Montréal, QCH2W 1R7, Canada
| | - Paul Le Tissier
- Centre for Integrative Physiology, University of Edinburgh, EdinburghEH8 9XD, United Kingdom
| | - Patrice Mollard
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, MontpellierF-34094, France
- BioCampus Montpellier, University of Montpellier, CNRS, INSERM, MontpellierF-34094, France
| | - Helen Christian
- Department of Physiology, Anatomy and Genetics, Oxford University, OxfordOX1 3QX, United Kingdom
| | - Jacques Drouin
- Laboratoire de génétique moléculaire, Institut de recherches cliniques de Montréal, Montréal, QCH2W 1R7, Canada
| |
Collapse
|
2
|
Kobayashi K, Yamamoto K, Kikuyama S, Tanaami Y, Machida T, Kobayashi T. Delayed Postnatal Growth and Anterior Pituitary Development in Growth-Retarded ( grt) Female Mice. Zoolog Sci 2021; 38:238-246. [PMID: 34057348 DOI: 10.2108/zs200063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 11/26/2020] [Indexed: 11/17/2022]
Abstract
Growth-retarded (grt) mice display primary congenital hypothyroidism due to the hyporesponsiveness of their thyroid glands to thyroid-stimulating hormone (TSH). We examined somatic growth, anterior pituitary development, and hormonal profiles in female grt mice and normal ones. Although growth in grt females was suppressed 2 weeks after birth, the measured growth parameters and organ weights gradually increased and finally reached close to the normal levels. Grt mice exhibited delayed eye and vaginal openings and remained in a state of persistent diestrus thereafter, plasma estrogen levels being lower than those in normal mice. Grt mice that received normal-donor thyroids showed accelerated growth and their body weights increased up to the sham-normal levels, indicating the importance of early thyroid hormone supplementation. In the anterior pituitary, there were fewer growth hormone (GH) and prolactin (PRL) cells in grt mice than in normal mice as examined at 12 weeks after birth, but the numbers of these cells did not differ from those in normal mice after 24 weeks. Grt mice had more TSH cells than normal mice until 48 weeks. Plasma GH levels in grt mice were lower than those in normal mice at 2 weeks, but did not differ substantially after 5 weeks. Compared with normal mice, grt mice had significantly lower plasma PRL and thyroxine levels, but notably higher TSH levels until 48 weeks. These findings indicate that thyroid hormone deficiency in grt mice causes delayed development and growth, and inappropriate development of GH, PRL and TSH cells, followed by the abnormal secretion of hormones by these pituitary cells.
Collapse
Affiliation(s)
- Kenichi Kobayashi
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan, .,National Institute of Occupational Safety and Health, Kawasaki 214-8585, Japan
| | - Kazutoshi Yamamoto
- Department of Biology, Faculty of Education and Integrated Sciences, Waseda University, Tokyo 162-8480, Japan
| | - Sakae Kikuyama
- Department of Biology, Faculty of Education and Integrated Sciences, Waseda University, Tokyo 162-8480, Japan
| | - Yukinobu Tanaami
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| | - Takeo Machida
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| | - Tetsuya Kobayashi
- Division of Life Science, Graduate School of Science and Engineering, Saitama University, Saitama 338-8570, Japan
| |
Collapse
|
3
|
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: 0.7] [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
|
4
|
Kapali J, Kabat BE, Schmidt KL, Stallings CE, Tippy M, Jung DO, Edwards BS, Nantie LB, Raeztman LT, Navratil AM, Ellsworth BS. Foxo1 Is Required for Normal Somatotrope Differentiation. Endocrinology 2016; 157:4351-4363. [PMID: 27631552 PMCID: PMC5086538 DOI: 10.1210/en.2016-1372] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The etiology for half of congenital hypopituitarism cases is unknown. Our long-term goal is to expand the molecular diagnoses for congenital hypopituitarism by identifying genes that contribute to this condition. We have previously shown that the forkhead box transcription factor, FOXO1, is present in approximately half of somatotropes at embryonic day (e) 18.5, suggesting it may have a role in somatotrope differentiation or function. To elucidate the role of FOXO1 in somatotrope differentiation and function, Foxo1 was conditionally deleted from the anterior pituitary (Foxo1Δpit). Uncommitted progenitor cells are maintained and able to commit to the somatotrope lineage normally based on the expression patterns of Sox2, a marker of uncommitted pituitary progenitors, and Pou1f1 (also known as Pit1), which marks committed progenitors. Interestingly, Foxo1Δpit embryonic mice exhibit delayed somatotrope differentiation as evidenced by an almost complete absence of GH immunoreactivity at e16.5 and reduced expression of Gh at e18.5 and postnatal day (P) 3. Consistent with this conclusion, expression of GHRH receptor, a marker of terminally differentiated somatotropes, is significantly reduced at e18.5 and P3 in the absence of FOXO1. The mechanism of FOXO1 regulation of somatotrope differentiation may involve the basic helix-loop-helix transcription factor, Neurod4, which has been implicated in somatotrope differentiation and is significantly reduced in Foxo1Δpit mice. Foxo1Δpit mice do not exhibit growth defects, and at P21 their pituitary glands exhibit a normal distribution of somatotropes. These studies demonstrate that FOXO1 is important for initial somatotrope specification embryonically but is dispensable for postnatal somatotrope expansion and growth.
Collapse
Affiliation(s)
- Jyoti Kapali
- Department of Physiology (J.K., B.E.K., K.L.S., C.E.S., M.T., D.O.J., B.S.El.), Southern Illinois University, Carbondale, Illinois 62901-6523; Department of Zoology and Physiology (B.S.Ed., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; and Department of Molecular and Integrative Physiology (L.B.N., L.T.R.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | - Brock E Kabat
- Department of Physiology (J.K., B.E.K., K.L.S., C.E.S., M.T., D.O.J., B.S.El.), Southern Illinois University, Carbondale, Illinois 62901-6523; Department of Zoology and Physiology (B.S.Ed., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; and Department of Molecular and Integrative Physiology (L.B.N., L.T.R.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | - Kelly L Schmidt
- Department of Physiology (J.K., B.E.K., K.L.S., C.E.S., M.T., D.O.J., B.S.El.), Southern Illinois University, Carbondale, Illinois 62901-6523; Department of Zoology and Physiology (B.S.Ed., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; and Department of Molecular and Integrative Physiology (L.B.N., L.T.R.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | - Caitlin E Stallings
- Department of Physiology (J.K., B.E.K., K.L.S., C.E.S., M.T., D.O.J., B.S.El.), Southern Illinois University, Carbondale, Illinois 62901-6523; Department of Zoology and Physiology (B.S.Ed., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; and Department of Molecular and Integrative Physiology (L.B.N., L.T.R.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | - Mason Tippy
- Department of Physiology (J.K., B.E.K., K.L.S., C.E.S., M.T., D.O.J., B.S.El.), Southern Illinois University, Carbondale, Illinois 62901-6523; Department of Zoology and Physiology (B.S.Ed., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; and Department of Molecular and Integrative Physiology (L.B.N., L.T.R.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | - Deborah O Jung
- Department of Physiology (J.K., B.E.K., K.L.S., C.E.S., M.T., D.O.J., B.S.El.), Southern Illinois University, Carbondale, Illinois 62901-6523; Department of Zoology and Physiology (B.S.Ed., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; and Department of Molecular and Integrative Physiology (L.B.N., L.T.R.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | - Brian S Edwards
- Department of Physiology (J.K., B.E.K., K.L.S., C.E.S., M.T., D.O.J., B.S.El.), Southern Illinois University, Carbondale, Illinois 62901-6523; Department of Zoology and Physiology (B.S.Ed., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; and Department of Molecular and Integrative Physiology (L.B.N., L.T.R.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | - Leah B Nantie
- Department of Physiology (J.K., B.E.K., K.L.S., C.E.S., M.T., D.O.J., B.S.El.), Southern Illinois University, Carbondale, Illinois 62901-6523; Department of Zoology and Physiology (B.S.Ed., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; and Department of Molecular and Integrative Physiology (L.B.N., L.T.R.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | - Lori T Raeztman
- Department of Physiology (J.K., B.E.K., K.L.S., C.E.S., M.T., D.O.J., B.S.El.), Southern Illinois University, Carbondale, Illinois 62901-6523; Department of Zoology and Physiology (B.S.Ed., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; and Department of Molecular and Integrative Physiology (L.B.N., L.T.R.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | - Amy M Navratil
- Department of Physiology (J.K., B.E.K., K.L.S., C.E.S., M.T., D.O.J., B.S.El.), Southern Illinois University, Carbondale, Illinois 62901-6523; Department of Zoology and Physiology (B.S.Ed., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; and Department of Molecular and Integrative Physiology (L.B.N., L.T.R.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| | - Buffy S Ellsworth
- Department of Physiology (J.K., B.E.K., K.L.S., C.E.S., M.T., D.O.J., B.S.El.), Southern Illinois University, Carbondale, Illinois 62901-6523; Department of Zoology and Physiology (B.S.Ed., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; and Department of Molecular and Integrative Physiology (L.B.N., L.T.R.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801
| |
Collapse
|
5
|
Clinical Outcome of Silent Subtype III Pituitary Adenomas Diagnosed by Immunohistochemistry. J Neuropathol Exp Neurol 2016; 74:1170-7. [PMID: 26574671 DOI: 10.1097/nen.0000000000000265] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Silent subtype III pituitary adenomas (SS-3) are nonfunctioning radiosensitive adenomas that may be associated with an increased risk of recurrence and invasion. The features that have been proposed to be diagnostically important are identifiable by electron microscopy (EM) and include an enlarged Golgi apparatus, along with several other ultrastructural features. The often limited availability of EM and the uncertainty about the relative importance of individual features pose practical challenges to the diagnosis. We hypothesized that it may be possible to diagnose SS-3 based solely on a markedly enlarged Golgi apparatus identified at the light microscopic level. In this prospective study, we used immunohistochemistry (IHC) for the Golgi apparatus with the MG-160/GLG-1 antibody to identify 10 cases with features suggestive of SS-3. Electron microscopy was performed for confirmation on 1 case. Compared with a control group of 20 conventional null cell adenomas, the SS-3 adenomas showed an increased MIB-1 proliferation index (p < 0.01), a higher risk of invasion (p < 0.01), and a higher incidence of recurrence (p < 0.01). Thus, in this first controlled study, we demonstrate that SS-3 is clinically aggressive and identifiable by IHC, without the need for EM. The routine diagnostic workup of nonsecreting adenomas should rule out SS-3, which can be done quickly and efficiently by IHC.
Collapse
|
6
|
Richardson TE, Mathis DA, Mickey BE, Raisanen JM, Burns DK, White CL, Hatanpaa KJ. Clinical Outcome of Silent Subtype III Pituitary Adenomas Diagnosed by Immunohistochemistry. J Neuropathol Exp Neurol 2015. [DOI: 10.1093/jnen/74.12.1170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
7
|
Wang W, Xu Z, Fu L, Liu W, Li X. Pathogenesis analysis of pituitary adenoma based on gene expression profiling. Oncol Lett 2014; 8:2423-2430. [PMID: 25360166 PMCID: PMC4214395 DOI: 10.3892/ol.2014.2613] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 09/08/2014] [Indexed: 12/21/2022] Open
Abstract
The aim of the current study was to investigate the pathogenesis of pituitary adenoma through screening of the differentially-expressed genes (DEGs) and proteins in normal pituitary and pituitary adenoma tissues, and analyzing the interactions among them. Following the acquisition of gene expression profiling data from a public functional genomics data repository, Gene Expression Omnibus, DEGs were screened in normal pituitary and pituitary adenoma tissues. Upregulated and downregulated DEGs were further identified through gene ontology functional enrichment analysis. Subsequently, the DEGs were mapped to the Search Tool for the Retrieval of Interacting Genes database, and the protein-protein interaction (PPI) networks of the upregulated and downregulated DEGs were constructed. Finally, the functional modules of the PPI network of the downregulated DEGs were analyzed. In total, 211 upregulated and 413 downregulated DEGs were screened between the normal pituitary and pituitary adenoma samples. Downregulated DEGs were associated with certain functions, including the immune response, hormone regulation and cell proliferation. Upregulated genes were associated with cation transport functions. Five modules were acquired from the PPI network of the downregulated DEGs. Transcription factors, including signal transducer and activator of transcription 3 (STAT3), interleukin 6 (IL-6), B-cell lymphoma 6 protein, early growth response 1, POU1F1, jun B proto-oncogene and FOS were the core nodes in the functional modules. In summary, the DEGs and proteins were identified through screening gene expression profiling and PPI networks. The results of the present study indicated that low expression levels of hormone- and immune-related genes facilitated the occurrence of pituitary adenoma. Low expression levels of IL-6 and STAT3 were significant in the dysimmunity of pituitary adenoma. Furthermore, the low expression level of POU1F1 contributed to the reduction in pituitary hormone secretion.
Collapse
Affiliation(s)
- Weimin Wang
- School of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China ; Department of Neurosurgery, Qingdao Municipal Hospital, Qingdao, Shandong 266071, P.R. China
| | - Zhiming Xu
- Department of Neurosurgery, Qingdao Municipal Hospital, Qingdao, Shandong 266071, P.R. China
| | - Li Fu
- Department of General Surgery, Qingdao Municipal Hospital, Qingdao, Shandong 266071, P.R. China
| | - Wei Liu
- Department of Neurosurgery, Qingdao Municipal Hospital, Qingdao, Shandong 266071, P.R. China
| | - Xingang Li
- School of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China ; Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
8
|
Abstract
Pituitary adenomas are classified by function as defined by clinical symptoms and signs of hormone hypersecretion with subsequent confirmation on immunohistochemical staining. However, positive immunostaining for pituitary cell types has been shown for clinically nonfunctioning adenomas, and this entity is classified as silent functioning adenoma. Most common in these subtypes include silent gonadotroph adenomas, silent corticotroph adenomas and silent somatotroph adenomas. Less commonly, silent prolactinomas and thyrotrophinomas are encountered. Appropriate classification of these adenomas may affect follow-up care after surgical resection. Some silent adenomas such as silent corticotroph adenomas follow a more aggressive course, necessitating closer surveillance. Furthermore, knowledge of the immunostaining characteristics of silent adenomas may determine postoperative medical therapy. This article reviews the incidence, clinical behavior, and pathologic features of clinically silent pituitary adenomas.
Collapse
Affiliation(s)
- Odelia Cooper
- Pituitary Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Shlomo Melmed
- Pituitary Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| |
Collapse
|
9
|
Castinetti F, Brinkmeier ML, Gordon DF, Vella KR, Kerr JM, Mortensen AH, Hollenberg A, Brue T, Ridgway EC, Camper SA. PITX2 AND PITX1 regulate thyrotroph function and response to hypothyroidism. Mol Endocrinol 2011; 25:1950-60. [PMID: 21964592 PMCID: PMC3386545 DOI: 10.1210/me.2010-0388] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Accepted: 08/30/2011] [Indexed: 01/05/2023] Open
Abstract
Pitx2 is a homeodomain transcription factor required in a dose-dependent manner for the development of multiple organs. Pitx2-null homozygotes (Pitx2(-/-)) have severe pituitary hypoplasia, whereas mice with reduced-function alleles (Pitx2(neo/neo)) exhibit modest hypoplasia and reduction in the developing gonadotroph and Pou1f1 lineages. PITX2 is expressed broadly in Rathke's pouch and the fetal pituitary gland. It predominates in adult thyrotrophs and gonadotrophs, although it is not necessary for gonadotroph function. To test the role of PITX2 in thyrotroph function, we developed thyrotroph-specific cre transgenic mice, Tg(Tshb-cre) with a recombineered Tshb bacterial artificial chromosome that ablates floxed genes in differentiated pituitary thyrotrophs. We used the best Tg(Tshb-Cre) strain to generate thyrotroph-specific Pitx2-deficient offspring, Pitx2(flox/-;)Tg(Tshb-cre). Double immunohistochemistry confirmed Pitx2 deletion. Pitx2(flox/-);Tg(Tshb-cre) mice have a modest weight decrease. The thyroid glands are smaller, although circulating T(4) and TSH levels are in the normal range. The pituitary levels of Pitx1 transcripts are significantly increased, suggesting a compensatory mechanism. Hypothyroidism induced by low-iodine diet and oral propylthiouracil revealed a blunted TSH response in Pitx2(flox/-);Tg(Tshb-cre) mice. Pitx1 transcripts increased significantly in control mice with induced hypothyroidism, but they remained unchanged in Pitx2(flox/-);Tg(Tshb-cre) mice, possibly because Pitx1 levels were already maximally elevated in untreated mutants. These results suggest that PITX2 and PITX1 have overlapping roles in thyrotroph function and response to hypothyroidism. The novel cre transgene that we report will be useful for studying the function of other genes in thyrotrophs.
Collapse
Affiliation(s)
- F Castinetti
- Department of Human Genetics, University of Michigan, Ann Arbor, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Goldberg LB, Aujla PK, Raetzman LT. Persistent expression of activated Notch inhibits corticotrope and melanotrope differentiation and results in dysfunction of the HPA axis. Dev Biol 2011; 358:23-32. [PMID: 21781958 DOI: 10.1016/j.ydbio.2011.07.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Revised: 06/17/2011] [Accepted: 07/05/2011] [Indexed: 11/30/2022]
Abstract
The hypothalamic-pituitary-adrenal (HPA) axis is an important regulator of energy balance, immune function and the body's response to stress. Signaling networks governing the initial specification of corticotropes, a major component of this axis, are not fully understood. Loss of function studies indicate that Notch signaling may be necessary to repress premature differentiation of corticotropes and to promote proliferation of pituitary progenitors. To elucidate whether Notch signaling must be suppressed in order for corticotrope differentiation to proceed and whether Notch signaling is sufficient to promote corticotrope proliferation, we examined the effects of persistent Notch expression in Pomc lineage cells. We show that constitutive activation of the Notch cascade inhibits the differentiation of both corticotropes and melanotropes and results in the suppression of transcription factors required for Pomc expression. Furthermore, persistent Notch signaling traps cells in the intermediate lobe of the pituitary in a progenitor state, but has no effect on pituitary proliferation. Undifferentiated cells are eliminated in the first two postnatal weeks in these mice, resulting in a modest increase in CRH expression in the paraventricular nucleus, hypoplastic adrenal glands and decreased stress-induced corticosterone levels. Taken together, these findings show that Notch signaling is sufficient to prevent corticotrope and melanotrope differentiation, resulting in dysregulation of the HPA axis.
Collapse
Affiliation(s)
- Leah B Goldberg
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | | | | |
Collapse
|
11
|
Leonardi R, Farella M, Cobourne MT. An association between sella turcica bridging and dental transposition. Eur J Orthod 2011; 33:461-5. [DOI: 10.1093/ejo/cjq106] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
12
|
Cooper O, Ben-Shlomo A, Bonert V, Bannykh S, Mirocha J, Melmed S. Silent corticogonadotroph adenomas: clinical and cellular characteristics and long-term outcomes. HORMONES & CANCER 2010; 1:80-92. [PMID: 20717480 PMCID: PMC2921667 DOI: 10.1007/s12672-010-0014-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Silent corticotrophins adenomas (SCAs) are clinically silent and non-secreting but immunostain positively for ACTH. We hypothesize that SCAs comprise both corticotroph and gonadotroph characteristics. Cohort analysis from 1994-2008 with follow-up time ranging from 1-15 years in a tertiary referral center. We compared preoperative and postoperative clinical results and tumor cytogenesis in 25 SCAs and 84 nonfunctioning adenomas in 109 consecutive patients diagnosed pre-operatively with nonfunctioning pituitary adenomas. Clinical outcomes were radiologic and hormonal measures. Pathologic outcomes were expression of relevant pituitary hormones, tissue-specific transcription factors, and electron microscopy features. Preoperative SCA presentation was similar to that observed for nonfunctioning adenomas. However, SCAs recurred postoperatively at a median of 3 years vs. 8 years for nonfunctioning adenomas (p<0.0001). Fifty-four percent of patients with SCAs had new onset postoperative hypopituitarism vs. 17% of nonfunctioning adenomas (p<0.025). SCAs (n=18) were immunopositive for ACTH, cytoplasmic and nuclear SF-1, NeuroD1, DAX-1, and alpha-gonadotropin subunit, but Tpit negative, and co-expression of tumor ACTH with either SF-1 or LH was detected. In contrast, functional corticotroph adenomas (n=11) were immunopositive for ACTH, nuclear SF-1, NeuroD1, and Tpit, but negative for DAX-1, a gonadotroph cell transcription factor. Gonadotroph adenomas (n=23) were immunonegative for ACTH and Tpit but positive for nuclear SF-1, NeuroD1, and DAX-1. SCA electron microscopy demonstrated ultrastructural features consistent with corticotroph and gonadotroph cells. As SCAs exhibit features consistent with both corticotroph and gonadotroph cytologic origin, we propose a pathologic and clinically distinct classification of SCAs as silent corticogonadotroph adenomas.
Collapse
Affiliation(s)
- Odelia Cooper
- Pituitary Center, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Room 2015, Los Angeles, CA 90048 USA
| | - Anat Ben-Shlomo
- Pituitary Center, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Room 2015, Los Angeles, CA 90048 USA
| | - Vivien Bonert
- Pituitary Center, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Room 2015, Los Angeles, CA 90048 USA
| | - Serguei Bannykh
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - James Mirocha
- Biostatistics Research Instititute, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - Shlomo Melmed
- Pituitary Center, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Room 2015, Los Angeles, CA 90048 USA
| |
Collapse
|
13
|
Life and death in aluminium-exposed cultures of rat lactotrophs studied by flow cytometry. Cell Biol Toxicol 2010; 26:341-53. [DOI: 10.1007/s10565-009-9147-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Accepted: 12/09/2009] [Indexed: 10/20/2022]
|
14
|
Monahan P, Rybak S, Raetzman LT. The notch target gene HES1 regulates cell cycle inhibitor expression in the developing pituitary. Endocrinology 2009; 150:4386-94. [PMID: 19541765 PMCID: PMC2736073 DOI: 10.1210/en.2009-0206] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The pituitary is an endocrine gland responsible for the release of hormones, which regulate growth, metabolism, and reproduction. Diseases such as hypopituitarism or pituitary adenomas are able to disrupt pituitary function leading to suboptimal function of the entire endocrine system. Growth of the pituitary during development and adulthood is a tightly regulated process. Hairy and enhancer of split (HES1), a transcription factor whose expression is initiated by the Notch signaling pathway, is a repressor of cell cycle inhibitors. We hypothesize that with the loss of Hes1, pituitary progenitors are no longer maintained in a proliferative state, choosing instead to exit the cell cycle. To test this hypothesis, we examined the expression of cell cycle regulators in wild-type and Hes1-deficient pituitaries. Our studies indicate that in early pituitary development [embryonic day (e) 10.5], cells contained in the Rathke's pouch of Hes1 mutants have decreased proliferation, indicated by changes in phosphohistone H3 expression. Furthermore, pituitaries lacking Hes1 have increased cell cycle exit, shown by significant increases in the cyclin-dependent kinase inhibitors, p27 and p57, from e10.5 to e14.5. Additionally, Hes1 mutant pituitaries have ectopic expression of p21 in Rathke's pouch progenitors, an area coincident with increased cell death. These observations taken together indicate a role for HES1 in the control of cell cycle exit and in mediating the balance between proliferation and differentiation, allowing for the properly timed emergence of hormone secreting cell types.
Collapse
Affiliation(s)
- Pamela Monahan
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | | | | |
Collapse
|
15
|
Abstract
Although pituitary tumors are mostly benign, they share certain molecular events with more malignant neoplasia, although their precise pathogenesis is far from established. The acquisition of new functional characteristics during their evolution suggests a multistep process that leads to tumor transformation. Mutations in classical tumor suppressor genes or oncogenes are infrequently associated with pituitary tumorigenesis. However, alterations in different signaling pathways, especially those involved in pituitary gland development, have emerged as significant features in pituitary adenomas. In particular, changes in inhibitory components of the beta-catenin pathway and its relationship to the cadherin family of peptides may well play an important role in tumorigenesis. We review and assess the role of the beta-catenin signaling pathway in the pathogenesis of pituitary adenomas.
Collapse
Affiliation(s)
- Maria Gueorguiev
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, Charterhouse Square, London EC1M 6BQ, UK
| | | |
Collapse
|
16
|
Himes AD, Raetzman LT. Premature differentiation and aberrant movement of pituitary cells lacking both Hes1 and Prop1. Dev Biol 2009; 325:151-61. [PMID: 18996108 PMCID: PMC2642967 DOI: 10.1016/j.ydbio.2008.10.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2008] [Revised: 09/26/2008] [Accepted: 10/03/2008] [Indexed: 12/24/2022]
Abstract
In the pituitary, the transition from proliferating progenitor cell into differentiated hormone producing cell is carefully regulated in a time-dependent and spatially-restricted manner. We report that two targets of Notch signaling, Hes1 and Prop1, are needed to maintain progenitors within Rathke's pouch and for the restriction of differentiated cells to the ventral pituitary. We observed ACTH and alphaGSU producing cells that had prematurely differentiated within Rathke's pouch along with correlated ectopic expression of Mash1 only when both Prop1 and Hes1 were lost. We also discovered that downregulation of N-cadherin expression in cells as they transition from Rathke's pouch to the anterior lobe appears to be essential for their movement. In the Prop1 mutant, cells are trapped in Rathke's pouch and N-cadherin expression remains high. Also, Slug, a marker of epithelial-to-mesenchymal transition, is absent in the dorsal anterior lobe. When Hes1 is lost in the Prop1 mutant, N-cadherin is downregulated and cells are able to exit Rathke's pouch but have lost their migrational cues and form ectopic foci surrounding Rathke's pouch. Our data reveal important overlapping functions of Hes1 and Prop1 in cell differentiation and movement that are critical for pituitary organogenesis.
Collapse
|
17
|
Charles MA, Mortensen AH, Potok MA, Camper SA. Pitx2 deletion in pituitary gonadotropes is compatible with gonadal development, puberty, and fertility. Genesis 2008; 46:507-14. [PMID: 18802953 PMCID: PMC2923441 DOI: 10.1002/dvg.20398] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
This report introduces a gonadotrope-specific cre transgenic mouse capable of ablating floxed genes in mature pituitary gonadotropes. Initial analysis of this transgenic line, Tg(Lhb-cre)1Sac, reveals that expression is limited to the pituitary cells that produce luteinizing hormone beta, beginning appropriately at e17.5. Cre activity is detectable by a reporter gene in nearly every LHbeta-producing cell, but the remaining hormone-producing cell types and other organs exhibit little to no activity. We used the Tg(Lhb-cre)1Sac strain to assess the role Pitx2 in gonadotrope function. The gonadotrope-specific Pitx2 knockout mice exhibit normal expression of LHbeta, sexual maturation, and fertility, suggesting that Pitx2 is not required for gonadotrope maintenance or for regulated production of gonadotropins.
Collapse
Affiliation(s)
- Michael A Charles
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan 48109-0618, USA
| | | | | | | |
Collapse
|
18
|
Million Passe CM, White CR, King MW, Quirk PL, Iovanna JL, Quirk CC. Loss of the protein NUPR1 (p8) leads to delayed LHB expression, delayed ovarian maturation, and testicular development of a sertoli-cell-only syndrome-like phenotype in mice. Biol Reprod 2008; 79:598-607. [PMID: 18495683 DOI: 10.1095/biolreprod.108.068304] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The high mobility group factor NUPR1, also known as p8 and com1, plays a role in temporal expression of the beta subunit of luteinizing hormone, LHB, during gonadotroph development. At Embryonic Day (e) 16.5, LHB is detectable in wild-type (Nupr1(+/+)) but not Nupr1 knockout (Nupr1(-/-)) mice. LHB is initiated by e17.5 in Nupr1(-/-) mice, and expression is fully recovered by Postnatal Day (p) 2. Factors indicative of pituitary maturation, GATA2, CGA, and TSH, are not differentially expressed in Nupr1(-/-) and Nupr1(+/+) embryos at e17.5. Therefore, the delay in LHB expression does not appear to result from delayed pituitary development. In addition, the role of NUPR1 in gonadotropin expression appears specific for LHB, as no difference in FSHB is observed in Nupr1(-/-) and Nupr1(+/+) embryos. The gonads are also impacted by the absence of NUPR1. Ovaries of female Nupr1(-/-) mice lack corpora lutea (CL) at 8 wk, an age at which CL are present in all Nupr1(+/+) littermates. Sexual maturity is recovered by 11 wk in Nupr1(-/-) mice. Conversely, the testes of Nupr1(-/-) males appear normal through 8 mo of age. By 10 mo, however, these mice develop a condition in which a significant number of seminiferous tubules lack germ cells, an abnormality reminiscent of human Sertoli-cell-only syndrome. NUPR1 is undetectable in Nupr1(+/+) gonadotrophs by p2 and remains absent in adulthood, but quantitative PCR analysis indicates Nupr1(+/+) adult ovaries and testes express Nupr1 mRNA. Therefore, the ovarian and testicular phenotypes may be due to the loss of NUPR1 directly at the gonads.
Collapse
|
19
|
Pals K, Roudbaraki M, Denef C. Growth hormone-releasing hormone and glucocorticoids determine the balance between luteinising hormone (LH) beta- and LH beta/follicle-stimulating hormone beta-positive gonadotrophs and somatotrophs in the 14-day-old rat pituitary tissue in aggregate cell culture. J Neuroendocrinol 2008; 20:535-48. [PMID: 18363807 DOI: 10.1111/j.1365-2826.2008.01698.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Fourteen-day-old rat pituitary tissue represents an attractive model for studying cell population dynamics, particularly of gonadotrophs. Prolonged three-dimensional culture in serum- and hormone-free medium causes a striking decline in somatotroph abundance but a several-fold rise in monohormonal LH beta-positive cell number, whereas bihormonal gonadotrophs almost disappear. In the present study, we investigated whether these changes are inter-related by examining the effects of growth hormone-releasing hormone (GHRH) and glucocorticoids, two protagonist regulators of somatotrophs. Cells were identified by single cell reverse transcriptase-polymerase chain reaction (RT-PCR) and immunofluorescence. Supplementation of the cultures for 2 weeks with GHRH (1 nm) did not augment the proportion of somatotrophs, but expanded the nonhormonal cell population. GHRH reduced the proportion of monohormonal luteinising hormone (LH)beta mRNA positive cells to approximately 50% of control, although the effect was not seen when these cells were visualised by immunostaining. Supplementation of the cultures with dexamethasone (4 nM) for 3 weeks partially rescued LH beta/follicle-stimulating hormone beta cells and fully rescued the GH mRNA cells in parallel with a decline in nonhormonal cell abundance, but strongly reduced bromodeoxyuridine labelling of GH-immunoreactive cells. As studied by patch-clamp single cell RT-PCR at the start of culture, GHRH caused an acute rise in intracellular [Ca(2+)] in some monohormonal GH cells, but at a higher incidence in cells expressing LH beta mRNA, alone or in combination with GH mRNA and/or pro-opiomelanocortin (POMC) mRNA. The present data suggest that, in the 14-day-old rat pituitary, the majority of GHRH target cells are cells expressing LH beta mRNA alone or in combination with GH and/or POMC mRNA. The data show co-regulation of gonadotroph and somatotroph population sizes by glucocorticoids and GHRH, with the former preserving bihormonal gonadotrophs and the latter repressing LH beta-only cell abundance. GHRH may not expand the somatotroph population unless glucocorticoid hormone is present to maintain terminal differentiation.
Collapse
Affiliation(s)
- K Pals
- Laboratory of Cell Pharmacology, University of Leuven, Medical School, Campus Gasthuisberg (O&N), Leuven, Belgium
| | | | | |
Collapse
|
20
|
Yin Z, Williams-Simons L, Rawahneh L, Asa S, Kirschner LS. Development of a pituitary-specific cre line targeted to the Pit-1 lineage. Genesis 2008; 46:37-42. [PMID: 18196598 DOI: 10.1002/dvg.20362] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Tissue-specific expression of the Cre recombinase is a well-established genetic tool to analyze gene function in specific tissues and cell types. In this report, we describe the generation of a new transgenic line that expresses Cre under the control of the rat growth hormone releasing hormone receptor (rGhrhr) promoter. This promoter, chosen to target the anterior pituitary, drives cre-mediated recombination in cells of the Pit1 lineage, including somatotrophs, lactotrophs, and thyrotrophs. Cre activity is first detected at embryonic day 13.5, and gradually increases to reach high level expression by postnatal day 2. In addition to the pituitary, rGhrhr-cre expression was detected in vibrissae and in hair follicles of the proximal limb, but not in other tissues. The rGhrhr-cre line will be a valuable tool for the study of the development of the pituitary Pit1 lineage and for the study of tumorigenesis involving these cells.
Collapse
Affiliation(s)
- Zhirong Yin
- Department of Molecular Virology, Immunology, and Molecular Genetics, The Ohio State University, Columbus, Ohio, USA
| | | | | | | | | |
Collapse
|
21
|
Vankelecom H. Non-hormonal cell types in the pituitary candidating for stem cell. Semin Cell Dev Biol 2007; 18:559-70. [PMID: 17509912 DOI: 10.1016/j.semcdb.2007.04.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2006] [Revised: 02/22/2007] [Accepted: 04/10/2007] [Indexed: 12/17/2022]
Abstract
Hormone balances in the body are primarily governed by the hypothalamus-pituitary system. For its pivotal role, the pituitary gland relies on an assortment of different hormone-producing cell types, the proportions of which dynamically change in response to fluctuating endocrine demands. Mechanisms of pituitary cellular plasticity are at present far from understood, and may include proliferation and transdifferentiation of hormonal cells. Whether new cells also originate by recruitment from stem cells is unsettled, although this idea has frequently been proposed. Here, I will review these data by focusing on the non-hormonal cell types that have been advanced as candidates for the pituitary stem cell position.
Collapse
Affiliation(s)
- Hugo Vankelecom
- Laboratory of Cell Pharmacology, Department of Molecular Cell Biology, Campus Gasthuisberg O&N1, Herestraat 49, University of Leuven (K.U.Leuven), B-3000 Leuven, Belgium.
| |
Collapse
|
22
|
Weiss JM, Stojilkovic SS, Diedrich K, Ortmann O. Effects of testosterone on hormonal content and calcium-dependent basal secretion in female rat pituitary cells. J Steroid Biochem Mol Biol 2007; 103:149-57. [PMID: 17084076 DOI: 10.1016/j.jsbmb.2006.09.038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2006] [Accepted: 09/14/2006] [Indexed: 10/24/2022]
Abstract
In vivo and in vitro effects of elevated androgens on agonist-induced gonadotropin secretion have been addressed previously. Here we investigated the effects of testosterone on hormonal content and basal (in the absence of agonists) hormone release in pituitary lactotrophs, somatotrophs and gonadotrophs from female rats. Furthermore we tested the hypothesis that testosterone action is dependent on the pattern of spontaneous and Bay K 8644 (a L-type calcium channel agonist) -induced calcium signalling. Mixed anterior pituitary cells were cultured in steroid containing or depleted media, and testosterone (1pM to 10nM) was added for 48h. Cells were studied for their spontaneous and Bay K 8644-induced calcium signalling pattern and total hormone levels (release and hormonal content). In lactotrophs, somatotrophs and gonadotrophs testosterone did not affect the pattern of spontaneous calcium signalling. Bay K 8644-induced calcium signalling and hormone release were not affected by testosterone. In both steroid-depleted and -containing medium, testosterone inhibited prolactin (PRL), luteinizing hormone (LH) and growth hormone (GH) cellular content and release in a dose-dependent manner, with IC(50)s in a sub-nanomolar concentration range. These results indicate that testosterone inhibits basal hormone release from lactotrophs, somatotrophs and gonadotrophs without affecting intracellular calcium signalling. This action of testosterone is not dependent on the presence of other steroid hormones.
Collapse
Affiliation(s)
- Juergen M Weiss
- Department of Obstetrics and Gynecology, Medical University Luebeck, Ratzeburger Allee 160, 23538 Luebeck, Germany.
| | | | | | | |
Collapse
|
23
|
Abstract
Tissue-specific stem cells are uncovered in a growing number of organs by their molecular expression profile and their potential for self-renewal, multipotent differentiation and tissue regeneration. Whether the pituitary gland also contains a pool of versatile 'master' cells that drive homeostatic, plastic and regenerative cell ontogenesis is at present unknown. Here, I will give an overview of data that may lend support to the existence of stem cells in the postnatal pituitary. During the many decades of pituitary research, various approaches have been used to hunt for the pituitary stem cells. Transplantation and regeneration studies advanced chromophobes as possible source of new hormonal cells. Clonogenicity approaches identified pituitary cells that clonally expand to floating spheres, or to colonies in adherent cell cultures. Behavioural characteristics and changes of marginal, follicular and folliculostellate cells during defined developmental and (patho-)physiological conditions have been interpreted as indicative of a stem cell role. Expression of potential stem cell markers like nestin, as well as topographical localization in the marginal zone around the cleft has also been considered to designate pituitary stem cells. Finally, a 'side population' was recently identified in the postnatal pituitary which in many other tissues represents a stem cell-enriched fraction. Taken together, in the course of the long-standing study of the pituitary, several arguments have been presented to support the existence of stem cells, and multiple cell types have been placed in the spotlight as possible candidates. However, none of these cells has until now unequivocally been shown to meet all quintessential characteristics of stem cells.
Collapse
Affiliation(s)
- Hugo Vankelecom
- Laboratory of Cell Pharmacology, Department of Molecular Cell Biology, University of Leuven, Leuven, Belgium.
| |
Collapse
|
24
|
Chen J, Crabbe A, Van Duppen V, Vankelecom H. The Notch Signaling System Is Present in the Postnatal Pituitary: Marked Expression and Regulatory Activity in the Newly Discovered Side Population. Mol Endocrinol 2006; 20:3293-307. [PMID: 16959876 DOI: 10.1210/me.2006-0293] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Recently, we discovered in the adult anterior pituitary a subset of cells with side population (SP) phenotype, enriched for expression of stem/progenitor cell-associated factors like Sca1, and of Notch1 and Hes (hairy and enhancer of split) 1, components of the classically developmental Notch pathway. In the present study, we elaborated the expression of the Notch signaling system in the postnatal pituitary, and examined its functional significance within the SP compartment. Using RT-PCR, we detected in the anterior pituitary of adult mouse the expression of all four vertebrate Notch receptors, as well as of Hes1, 5, and 6, key downstream targets and effectors of Notch. All Notch receptors, Hes1 and Hes5 were measured at higher mRNA levels in the Sca1(high) SP than in the main population (MP) of differentiated hormonal cells. In contrast, Hes6, known as an inhibitor of Hes1, was more abundant in the MP. Cells with SP phenotype, enriched for Sca1(high) expression, were detected throughout postnatal life. Their proportion was higher in immature mice, but did not change from adult (8 wk old) to much older age (1 yr old). Notch pathway expression was higher in the Sca1(high) SP than in the MP at all postnatal ages analyzed. Functional implication of Notch signaling in the SP was investigated in reaggregate cultures of adult mouse anterior pituitary cells. Treatment with the gamma-secretase inhibitor DAPT down-regulated Notch activity and reduced the proportion of SP cells. Activation of Notch signaling with the conserved DSL motif of Notch ligands, or with a soluble ligand, caused a rise in SP cell number, at least in part due to a proliferative effect. The SP also expanded in proportion when aggregates were treated with leukemia-inhibitory factor, basic fibroblast growth factor, and epidermal growth factor, again at least partly accounted for by a mitogenic action. These intrapituitary growth factors all activated Notch signaling, and DAPT abrogated the expansion of the SP by basic fibroblast growth factor and leukemia-inhibitory factor, thus exposing a possible cross talk. In conclusion, we show that the Notch pathway, typically situated in embryogenesis, is also present and active in the postnatal pituitary, that it is particularly expressed within the SP independent of age, and that it plays a role in the regulation of SP abundance. Whether our data indicate that Notch regulates renewal and fate decisions of putative stem/progenitor cells within the pituitary SP as found in other tissues, remains open for further exploration.
Collapse
Affiliation(s)
- Jianghai Chen
- Laboratory of Cell Pharmacology, Department of Molecular Cell Biology, University of Leuven (K.U.Leuven), Campus Gasthuisberg O&N1, Herestraat 49, B-3000 Leuven, Belgium
| | | | | | | |
Collapse
|
25
|
Raetzman LT, Cai JX, Camper SA. Hes1 is required for pituitary growth and melanotrope specification. Dev Biol 2006; 304:455-66. [PMID: 17367776 PMCID: PMC1913046 DOI: 10.1016/j.ydbio.2006.11.010] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2006] [Revised: 10/25/2006] [Accepted: 11/06/2006] [Indexed: 11/23/2022]
Abstract
Rathke's pouch contains progenitor cells that differentiate into the endocrine cells of the pituitary gland. It gives rise to gonadotrope, thyrotrope, somatotrope, corticotrope and lactotrope cells in the anterior lobe and the intermediate lobe melanotropes. Pituitary precursor cells express many members of the Notch signaling pathway including the downstream effector gene Hes1. We hypothesized that Hes1 regulates the timing of precursor differentiation and cell fate determination. To test this idea, we expressed Hes1 in differentiating pituitary cells and found that it can inhibit gonadotrope and thyrotrope differentiation. Pituitaries of Hes1 deficient mice have anterior lobe hypoplasia. All cells in the anterior lobe are specified and differentiate, but an early period of increased cell death and reduced proliferation causes reduced growth, evident as early as e14.5. In addition, cells within the intermediate lobe differentiate into somatotropes instead of melanotropes. Thus, the Hes1 repressor is essential for melanotrope specification. These results demonstrate that Notch signaling plays multiple roles in pituitary development, influencing precursor number, organ size, cell differentiation and ultimately cell fate.
Collapse
|
26
|
Denef C, Pals K, Hauspie A, Vankelecom H, Seuntjens E. Combinatorial expression of phenotypes of different cell lineages in the rat and mouse pituitary. Ann N Y Acad Sci 2006; 1040:84-8. [PMID: 15891010 DOI: 10.1196/annals.1327.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
As studied by single cell RT-PCR of pituitary hormones, we demonstrated that the pituitaries of rats and mice contain a subpopulation of cells that express two or more hormone phenotypes typically belonging to lineages that are branched separately early during embryonic development, such as glycoprotein hormone alpha-subunit (alphaGSU) mRNA + PRL mRNA, alphaGSU mRNA + POMC mRNA, and POMC mRNA + GH or PRL mRNA. GnRH in vitro selectively expands the population of cells coexpressing alphaGSU mRNA + PRL mRNA, and CRH selectively increases the proportion of cells coexpressing alphaGSU mRNA + POMC mRNA. Colocalization of alphaGSU + PRL or alphaGSU + POMC could not be detected by double immunofluorescence. This lineage promiscuity was also observed in the pituitary in vivo.
Collapse
Affiliation(s)
- C Denef
- Laboratory of Cell Pharmacology, University of Leuven, School of Medicine, Belgium.
| | | | | | | | | |
Collapse
|
27
|
López-Domínguez AM, Espinosa JL, Navarrete A, Avila G, Cota G. Nerve growth factor affects Ca2+ currents via the p75 receptor to enhance prolactin mRNA levels in GH3 rat pituitary cells. J Physiol 2006; 574:349-65. [PMID: 16690703 PMCID: PMC1817754 DOI: 10.1113/jphysiol.2006.110791] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
In clonal pituitary GH(3) cells, spontaneous action potentials drive the opening of Ca(v)1 (L-type) channels, leading to Ca(2+) transients that are coupled to prolactin gene transcription. Nerve growth factor (NGF) has been shown to stimulate prolactin synthesis by GH(3) cells, but the underlying mechanisms are unknown. Here we studied whether NGF influences prolactin gene expression and Ca(2+) currents. By using RT-PCR, NGF (50 ng ml(-1)) was found to augment prolactin mRNA levels by approximately 80% when applied to GH(3) cells for 3 days. A parallel change in the prolactin content was detected by Western blotting. Both NGF-induced responses were mimicked by an agonist (Bay K 8644) and prevented by a blocker (nimodipine) of L-type channels. In whole-cell patch-clamp experiments, NGF enhanced the L-type Ca(2+) current by approximately 2-fold within 60 min. This effect reversed quickly upon growth factor withdrawal, but was maintained for days in the continued presence of NGF. In addition, chronic treatment (>or= 24 h) with NGF amplified the T-type current, which flows through Ca(v)3 channels and is thought to support pacemaking activity. Thus, NGF probably increases the amount of Ca(2+) that enters per action potential and may also induce a late increase in spike frequency. MC192, a specific antibody for the p75 neurotrophin receptor, but not tyrosine kinase inhibitors (K252a and lavendustin A), blocked the effects of NGF on Ca(2+) currents. Overall, the results indicate that NGF activates the p75 receptor to cause a prolonged increase in Ca(2+) influx through L-type channels, which in turn up-regulates the prolactin mRNA.
Collapse
Affiliation(s)
- Adriana M López-Domínguez
- Department of Physiology, Biophysics and Neurosciences, Cinvestav-IPN, AP 14-740, Mexico, DF 07000, Mexico
| | | | | | | | | |
Collapse
|
28
|
Nica G, Herzog W, Sonntag C, Nowak M, Schwarz H, Zapata AG, Hammerschmidt M. Eya1 is required for lineage-specific differentiation, but not for cell survival in the zebrafish adenohypophysis. Dev Biol 2006; 292:189-204. [PMID: 16458879 DOI: 10.1016/j.ydbio.2005.12.036] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2005] [Revised: 12/19/2005] [Accepted: 12/20/2005] [Indexed: 11/23/2022]
Abstract
The homeodomain transcription factor Six1 and its modulator, the protein phosphatase Eya1, cooperate to promote cell differentiation and survival during mouse organ development. Here, we studied the effects caused by loss of eya1 and six1 function on pituitary development in zebrafish. eya1 and six1 are co-expressed in all adenohypophyseal cells. Nevertheless, eya1 (aal, dog) mutants show lineage-specific defects, defining corticotropes, melanotropes, and gonadotropes as an Eya1-dependent lineage, which is complementary to the Pit1 lineage. Furthermore, eya1 is required for maintenance of pit1 expression, leading to subsequent loss of cognate hormone gene expression in thyrotropes and somatotropes of mutant embryos, whereas prolactin expression in lactotropes persists. In contrast to other organs, adenohypophyseal cells of eya1 mutants do not become apoptotic, and the adenohypophysis remains at rather normal size. Also, cells do not trans-differentiate, as in the case of pit1 mutants, but display morphological features characteristic for nonsecretory cells. Some of the adenohypophyseal defects of eya1 mutants are moderately enhanced in combination with antisense-mediated loss of Six1 function, which per se does not affect pituitary cell differentiation. In conclusion, this is the first report of an essential role of Eya1 during pituitary development in vertebrates. Eya1 is required for lineage-specific differentiation of adenohypophyseal cells, but not for their survival, thereby uncoupling the differentiation-promoting and anti-apoptotic effects of Eya proteins seen in other tissues.
Collapse
Affiliation(s)
- Gabriela Nica
- Max-Planck Institute of Immunobiology, Stuebeweg 51, 79108 Freiburg, Germany
| | | | | | | | | | | | | |
Collapse
|
29
|
Ward RD, Stone BM, Raetzman LT, Camper SA. Cell proliferation and vascularization in mouse models of pituitary hormone deficiency. Mol Endocrinol 2006; 20:1378-90. [PMID: 16556738 DOI: 10.1210/me.2005-0409] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Mutations in the transcription factors PIT1 (pituitary transcription factor 1) and PROP1 (prophet of Pit1) lead to pituitary hormone deficiency and hypopituitarism in mice and humans. To determine the basis for this, we performed histological analysis of Pit1- and Prop1-deficient dwarf mouse pituitaries throughout fetal and postnatal development. Pit1-deficient mice first exhibit pituitary hypoplasia after birth, primarily caused by reduced cell proliferation, although there is some apoptosis. To determine whether altered development of the vascular system contributes to hypopituitarism, we examined vascularization from embryonic d 14.5 and throughout development. No obvious differences in vascularization are evident in developing Pit1-deficient pituitaries. In contrast, the Prop1-deficient mouse pituitaries are poorly vascularized and dysmorphic, with a striking elevation in apoptosis. At postnatal d 11, apoptosis-independent caspase-3 activation occurs in thyrotropes and somatotropes of normal but not mutant pituitaries. This suggests that Prop1 and/or Pit1 may be necessary for caspase-3 expression. These studies provide further insight as to the mechanisms of Prop1 and Pit1 action in mice.
Collapse
Affiliation(s)
- Robert D Ward
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor 48109-0618, USA
| | | | | | | |
Collapse
|
30
|
Horvath E, Kovacs K, Smyth HS, Cusimano M, Singer W. Silent adenoma subtype 3 of the pituitary--immunohistochemical and ultrastructural classification: a review of 29 cases. Ultrastruct Pathol 2006; 29:511-24. [PMID: 16316952 DOI: 10.1080/01913120500323514] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The silent adenoma subtype 3 (SAS-3) of undetermined cellular derivation is a seemingly nonfunctioning aggressive pituitary tumor with a high recurrence rate. At the time of diagnosis SAS-3s are macro- or giant adenomas particularly aggressive in young individuals, especially women. They are usually associated with mild hyperprolactinemia and are unremarkable by histology. Immunohistochemistry, demonstrating scattered immunoreactivity mostly for GH, PRL, TSH, and alpha-subunit, is not diagnostic. Presently, only TEM permits conclusive diagnosis. Ultrastructurally, the large polar adenoma cells contain abundant RER, masses of SER, extensive multipolar Golgi apparatus, and unevenly clustered mitochondria, displaced by RER and SER, which may show close spatial relationship to RER. Cell membranes often form plexiform interdigitations. Nuclear pleomorphism and nuclear inclusions are common. The 100- to 200-nm secretory granules accumulate heavily in cell processes, which is a hallmark of glycoprotein hormone cell differentiation. The endothelial cells may contain tubuloreticular inclusions. Complete surgical removal of the large often invasive tumors is difficult necessitating postoperative treatment. SAS-3 is sensitive to conventional radiation. Some tumors express somatostatin receptors and respond well to somatostatin analogues, offering long-term control in patients with residual tumor. Possible derivation of SAS-3 from rostral thyrotrophs, a cell type presently known in rodents is contemplated.
Collapse
Affiliation(s)
- E Horvath
- Department of Laboratory Medicine, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada.
| | | | | | | | | |
Collapse
|
31
|
Pals K, Vankelecom H, Denef C. Triiodothyronine expands the lactotroph and maintains the lactosomatotroph population, whereas thyrotrophin-releasing hormone augments thyrotroph abundance in aggregate cell cultures of postnatal rat pituitary gland. J Neuroendocrinol 2006; 18:203-16. [PMID: 16454804 DOI: 10.1111/j.1365-2826.2005.01404.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In the present study, we used a three-dimensional pituitary cell culture system from early postnatal rats to examine the in vitro developmental potential of triiodothyronine (T3) and thyrotrophin-releasing hormone (TRH). Cell types were identified at the hormone mRNA level by single-cell reverse transcription-polymerase chain reaction and any change in abundance was further examined by immunofluorescence staining of the corresponding hormone protein. In aggregates from 14-day-old rats, long-term (12-16 days) treatment with T3 (0.5 nM) increased the abundance of cells expressing prolactin mRNA (PRLmRNA cells) by 2.5-fold and lowered that of nonhormonal cells and thyroid-stimulating hormone beta (TSHbeta)mRNA cells. The abundance of growth hormone (GH)mRNA cells decreased during culture compared to that in the freshly dispersed pituitary gland and T3 did not significantly affect this cell population. Cells coexpressing PRL mRNA and GH mRNA virtually disappeared during culture but reappeared in the presence of T3. T3 increased the abundance of PRL-immunoreactive (ir) cells in aggregates from 14-day-old rats, as well as in aggregates from newborn and 1-week-old rats. As estimated by bromodeoxyuridine (BrdU) labelling, a 3-day treatment with T3 enhanced the number of PRL-ir cells that had incorporated BrdU, but did not yet expand the total population of PRL-ir cells. Long-term treatment with TRH (100 nM) did not affect the proportion of PRLmRNA or GHmRNA cells, but consistently increased the proportional number of TSHbeta(mRNA) and TSHbeta-ir cells. The present data confirm the findings obtained in recent in vivo loss of function genetic studies suggesting that T3 plays a prominent role in postnatal expansion of the lactotroph population and that TRH is important for thyrotroph development. The data suggest that the effect of T3 is brought about by a direct action on the pituitary gland through a cell proliferation mechanism. T3 also appears to support the lactosomatotroph population. In view of the established theory that lactotrophs develop from GH-expressing progenitor cells and that this is a post mitotic event, we propose that T3 is mitogenic for GHmRNA cells that lack GH-ir material and that transdifferentiate into PRL-ir cells, but that a pathway of PRL cell development from mitotic nonhormonal cell progenitors may also be involved.
Collapse
Affiliation(s)
- K Pals
- Laboratory of Cell Pharmacology, University of Leuven (K.U.Leuven), Medical School, Campus Gasthuisberg (O & N), B-3000, Leuven, Belgium
| | | | | |
Collapse
|
32
|
Pogoda HM, von der Hardt S, Herzog W, Kramer C, Schwarz H, Hammerschmidt M. The proneural gene ascl1a is required for endocrine differentiation and cell survival in the zebrafish adenohypophysis. Development 2006; 133:1079-89. [PMID: 16481349 DOI: 10.1242/dev.02296] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mammalian basic helix-loop-helix proteins of the achaete-scute family are proneural factors that, in addition to the central nervous system, are required for the differentiation of peripheral neurons and sensory cells, derivatives of the neural crest and placodal ectoderm. Here, in identifying the molecular nature of the pia mutation, we investigate the role of the zebrafish achaete-scute homologue ascl1a during development of the adenohypophysis, an endocrine derivative of the placodal ectoderm. Similar to mutants deficient in Fgf3 signaling from the adjacent ventral diencepahalon, pia mutants display failure of endocrine differentiation of all adenohypophyseal cell types. Shortly after the failed first phase of cell differentiation, the adenohypophysis of pia mutants displays a transient phase of cell death, which affects most, but not all adenohypophyseal cells. Surviving cells form a smaller pituitary rudiment, lack expression of specific adenohypophyseal marker genes (pit1, neurod), while expressing others (lim3, pitx3), and display an ultrastructure reminiscent of precursor cells. During normal development, ascl1a is expressed in the adenohypophysis and the adjacent diencephalon, the source of Fgf3 signals. However, chimera analyses show that ascl1a is required cell-autonomously in adenohypophyseal cells themselves. In fgf3 mutants, adenohypophyseal expression of ascl1a is absent, while implantation of Fgf3-soaked beads into pia mutants enhances ascl1a, but fails to rescue pit1 expression. Together, this suggests that Ascl1a might act downstream of diencephalic Fgf3 signaling to mediate some of the effects of Fgf3 on the developing adenohypophysis.
Collapse
|
33
|
Glucocorticoid-Inducible Glutamine Synthetase in GH Cells of Chick Embryos, Gallus domesticus: Ontogeny of Glutamine Synthetase, GH, and Pit-1 Protein in the Pituitary Gland. J Poult Sci 2006. [DOI: 10.2141/jpsa.43.266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
34
|
van Tijn DA, de Vijlder JJM, Verbeeten B, Verkerk PH, Vulsma T. Neonatal detection of congenital hypothyroidism of central origin. J Clin Endocrinol Metab 2005; 90:3350-9. [PMID: 15784706 DOI: 10.1210/jc.2004-2444] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Due to the high frequency of concurrent pituitary hormone deficiencies, congenital hypothyroidism (CH) of central origin (CH-C) is a life-threatening disorder. Yet only a minority of these patients are detected by neonatal CH screening programs worldwide. We conducted a prospective multicenter study involving a 2-yr cohort of neonatally diagnosed CH-C patients to determine whether a T(4)-TSH-based neonatal CH screening protocol extended with T(4) binding globulin determinations improves early detection of CH-C and to assess the extent of pituitary hormone deficiency among the identified CH-C patients. In all infants with screening results indicative of CH-C, the functional integrity of the hypothalamo-hypophyseal system was investigated by dynamic tests; the anatomical integrity was investigated by magnetic resonance imaging. Initial test results were evaluated after 5 yr of follow-up. Among 385,000 infants screened over the 2-yr period, 19 cases of permanent CH-C were detected (prevalence, 1:20,263; 95% confidence interval, 1:12,976 to 1:33,654), representing 13.5% of all detected cases of permanent CH. The majority (78%) had multiple pituitary hormone deficiency, whereas 53% had pituitary malformations on magnetic resonance imaging. We conclude that infants with CH-C can very well be detected by neonatal screening. The estimated prevalence and the severity of pituitary dysfunction of this treatable disorder call for explicit attention for this entity of CH in neonatal screening programs worldwide.
Collapse
Affiliation(s)
- David A van Tijn
- Department of Pediatric Endocrinology, Emma Children's Hospital AMC, Academic Medical Center, G2-133, University of Amsterdam, P.O. Box 22700, 1100 DE Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
35
|
Ooi GT, Tawadros N, Escalona RM. Pituitary cell lines and their endocrine applications. Mol Cell Endocrinol 2004; 228:1-21. [PMID: 15541569 DOI: 10.1016/j.mce.2004.07.018] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2004] [Accepted: 07/15/2004] [Indexed: 10/26/2022]
Abstract
The pituitary gland is an important component of the endocrine system, and together with the hypothalamus, exerts considerable influence over the functions of other endocrine glands. The hypothalamus either positively or negatively regulates hormonal productions in the pituitary through its release of various trophic hormones which act on specific cell types in the pituitary to secrete a variety of pituitary hormones that are important for growth and development, metabolism, reproductive and nervous system functions. The pituitary is divided into three sections-the anterior lobe which constitute the majority of the pituitary mass and is composed primarily of five hormone-producing cell types (thyrotropes, lactotropes, corticotropes, somatotropes and gonadotropes) each secreting thyrotropin, prolactin, ACTH, growth hormone and gonadotropins (FSH and LH) respectively. There is also a sixth cell type in the anterior lobe-the non-endocrine, agranular, folliculostellate cells. The intermediate lobe produces melanocyte-stimulating hormone and endorphins, whereas the posterior lobe secretes anti-diuretic hormone (vasopressin) and oxytocin. Representative cell lines of all the six cell types of the anterior pituitary have been established and have provided valuable information on genealogy of the various cell lineages, endocrine feedback control of hormone synthesis and secretions, intrapituitary interactions between the various cell types, as well as the role of specific transcription factors that determine each differentiated cell phenotype. In this review, we will discuss the morphology and function of the cell types that make up the anterior pituitary, and the characteristics of the various functional anterior pituitary cell systems that have been established to be representative of each anterior pituitary cell lineage.
Collapse
Affiliation(s)
- Guck T Ooi
- Prince Henry's Institute of Medical Research, Monash Medical Centre, Block E, Level 4, 246 Clayton Road, Clayton, Victoria 3168, Australia.
| | | | | |
Collapse
|
36
|
Ward RD, Raetzman LT, Suh H, Stone BM, Nasonkin IO, Camper SA. Role of PROP1 in pituitary gland growth. Mol Endocrinol 2004; 19:698-710. [PMID: 15591534 DOI: 10.1210/me.2004-0341] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Mutations in the PROP1 transcription factor gene lead to reduced production of thyrotropin, GH, prolactin, and gonadotropins as well as to pituitary hypoplasia in adult humans and mice. Some PROP1-deficient patients initially exhibit pituitary hyperplasia that resolves to hypoplasia. To understand this feature and to explore the mechanism whereby PROP1 regulates anterior pituitary gland growth, we carried out longitudinal studies in normal and Prop1-deficient dwarf mice from early embryogenesis through adulthood, examining the volume of Rathke's pouch and its derivatives, the position and number of dividing cells, the rate of apoptosis, and cell migration by pulse labeling. The results suggest that anterior pituitary progenitors normally leave the perilumenal region of Rathke's pouch and migrate to form the anterior lobe as they differentiate. Some of the cells that seed the anterior lobe during organogenesis have proliferative potential, supporting the expansion of the anterior lobe after birth. Prop1-deficient fetal pituitaries are dysmorphic because mutant cells are retained in the perilumenal area and fail to differentiate. After birth, mutant pituitaries exhibit enhanced apoptosis and reduced proliferation, apparently because the mutant anterior lobe is not seeded with progenitors. These studies suggest a mechanism for Prop1 action and an explanation for some of the clinical findings in human patients.
Collapse
Affiliation(s)
- Robert D Ward
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan 48109-0638, USA
| | | | | | | | | | | |
Collapse
|
37
|
Maseki Y, Nakamura K, Iwasawa A, Zheng J, Inoue K, Sakai T. Development of gonadotropes in the chicken embryonic pituitary gland. Zoolog Sci 2004; 21:435-44. [PMID: 15118231 DOI: 10.2108/zsj.21.435] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Although a number of immunohistochemical studies have been carried out on the differentiation of chicken gonadotropes during embryogenesis, the temporal and spatial properties of appearance of gonadotropes are not clear. In this study, we studied the appearance and morphological characteristics of gonadotropes in the embryonic and adult chicken anterior pituitary glands using RT-PCR, in situ hybridization and immunohistochemistry. For this purpose, we raised specific antisera against chicken follicle-stimulating hormone beta-subunit (cFSHbeta) and chicken luteinizing hormone beta-subunit (cLHbeta) based on each putative amino acid sequence. RT-PCR analysis revealed that cFSHbeta mRNA was expressed from embryonic day 7 (E7). Chicken FSHbeta mRNA-expressing (-ex) and -immunopositive (-ip) cells started to appear in the ventral part of the caudal lobe in the anterior pituitary gland at E8. Chicken LHbeta-ip cells were also first observed there at E8, but cLH mRNA expression was confirmed from E4 by RT-PCR analysis. The distribution of these chicken gonadotropin-ex and -ip cells spread from the ventral part to dorsal part in the caudal lobe around E10 and subsequently expanded to the cephalic lobe from E12 to E20. These cells were morphologically classified into two types (round- and club-shaped cells). It was found that the density of gonadotropin-ip cells in the caudal lobe was always higher than that in the cephalic lobe throughout the period of development. To the best of our knowledge, this is the first report focusing on the differentiation of chicken gonadotropes by assessment of both protein and mRNA of chicken gonadotropin.
Collapse
Affiliation(s)
- Yoko Maseki
- Department of Regulation-Biology, Faculty of Science, Saitama University, 255 Shimo-ohkubo, Saitama, Saitama 338-8570, Japan
| | | | | | | | | | | |
Collapse
|
38
|
Radian S, Coculescu M, Morris JF. Somatotroph to thyrotroph cell transdifferentiation during experimental hypothyroidism - a light and electron-microscopy study. J Cell Mol Med 2004; 7:297-306. [PMID: 14594554 PMCID: PMC6741402 DOI: 10.1111/j.1582-4934.2003.tb00230.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Somatotroph and thyrotroph pituitary cells share a common precursor cell expressing the transcription factor Pit1 in ontogeny. Cells expressing both thyrotropin (TSH) and growth-hormone (GH) are found in adult rat pituitary and in human pituitary adenomas in acromegaly, and these tumors contain both thyrotropin-releasing hormone (TRH) and the TRH receptors (TRHR). It has been shown that stimulation of TSH expression in primary hypothyroidism promotes changes suggestive of somatotroph to thyrotroph cell transdifferentiation. We tested this hypothesis and the role of TRH in experimental primary hypothyroidism in rats. Adult female Long-Evans rats, 6 months old, were administered the antithyroid drug methimazole (0.1% w/v) in the drinking water for 42 days. Animals were sacrificed by perfusion fixation under anaesthesia at weekly intervals and pituitary tissue processed in acrylic resin for immunofluorescence and immuno-electronmicroscopy for TSH, GH and TRHR. In the hypothyroid rat pituitary immunofluorescent somatotrophs were greatly reduced in number and gradually replaced by thyrotrophs during methimazole administration. Colocalization of GH and TSH in the same cell was noted. Immunoelectronmicroscopy demonstrated the development of enlarged thyrotrophs with dilated rough endoplasmic reticulum containing an electron-dense material and intracisternal granules, both of which are immunoreactive for TSH ('thyroidectomy cells'). The somatotrophs showed reduced GH immunoreactivity and also the presence of TSH-type, small-size secretory granules. This suggests that the greatly increased number of TSH-cells in methimazole-induced-hypothyroidism is due, at least partially, to the transdifferentiation of somatotroph into thyrotroph cells. TRHR immunofluorescence was expressed in many somatotrophs in normal rat pituitary and unlike immunoreactive GH, its expression was enhanced during hypothyroidism. The number of TRHR-immunoreactive cells increased in parallel with the number of TSH-immunoreactive cells. This indicates a role for TRH stimulation in the transdifferentiation process. Taken together, these data suggest that, in addition to the cell mutation mechanism involving an early totipotential progenitor cell, transdifferentiation of existing somatotroph cells also plays a part in the pathogenesis of multihormonal GH-secreting adenomas.
Collapse
Affiliation(s)
- S Radian
- Carol Davila University of Medicine and Pharmacy, Department of Endocrinology, Bucharest, Romania.
| | | | | |
Collapse
|
39
|
Herzog W, Sonntag C, Walderich B, Odenthal J, Maischein HM, Hammerschmidt M. Genetic analysis of adenohypophysis formation in zebrafish. Mol Endocrinol 2004; 18:1185-95. [PMID: 14752054 DOI: 10.1210/me.2003-0376] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The adenohypophysis consists of at least six different cell types, somatotropes, lactotropes, thyrotropes, melanotropes, corticotropes, and gonadotropes. In mouse, cloning of spontaneous mutations and gene targeting has revealed multiple genes required for different steps of adenohypophysis development. Here, we report the results of a systematic search for genes required for adenohypophysis formation and patterning in zebrafish. By screening F3 offspring of N-ethyl-N-nitrosourea-mutagenized founder fish, we isolated eleven mutants with absent or reduced expression of GH, the product of somatotropes, but a normally developing hypothalamus. Of such mutants, eight were further analyzed and mapped. They define four genes essential for different steps of adenohypophysis development. Two of them, lia and pia, affect the entire adenohypophysis, whereas the other two are required for a subset of adenohypophyseal cell types only. The third gene is zebrafish pit1 and is required for lactotropes, thyrotropes, and somatotropes, similar to its mouse ortholog, whereas the fourth, aal, is required for corticotropes, melanotropes, thyrotropes, and somatotropes, but not lactotropes. In conclusion, the isolated zebrafish mutants confirm principles of adenohypophysis development revealed in mouse, thereby demonstrating the high degree of molecular and mechanistic conservation among the different vertebrate species. In addition, they point to thus far unknown features of adenohypophysis development, such as the existence of a new lineage of pituitary cells, which partially overlaps with the Pit1 lineage. Positional cloning of the lia, pia, and aal genes might reveal novel regulators of vertebrate pituitary development.
Collapse
Affiliation(s)
- Wiebke Herzog
- Max-Planck Institute for Immunobiology, Stuebeweg 51, 79108 Freiburg, Germany
| | | | | | | | | | | |
Collapse
|
40
|
Ferretti E, Di Stefano D, Zazzeroni F, Gallo R, Fratticci A, Carfagnini R, Angiulli S, Santoro A, Minniti G, Tamburrano G, Alesse E, Cantore G, Gulino A, Jaffrain-Rea ML. Human pituitary tumours express the bHLH transcription factors NeuroD1 and ASH1. J Endocrinol Invest 2003; 26:957-65. [PMID: 14759067 DOI: 10.1007/bf03348192] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Among the transcription factors involved in pituitary ontogenesis and physiology, basic helix-loop-helix (bHLH) have been poorly studied. Members of bHLH family include NeuroD1 and ASH1, both involved in neuroendocrine differentiation. We evaluated their mRNA expression patterns, by semi-quantitative RT-PCR analysis (sq-RT-PCR) and/or Northern blot, in a series of 33 pituitary adenomas (PA), anterior pituitaries, and pituitary cell lines. Immunohistochemistry for NeuroD1 was also performed in 25 PA. Low levels of NeuroD1 were observed in normal pituitaries and in the somatomammotroph cell lines GH3/GH4C1, contrasting with high levels in corticotroph AtT20 cells. NeuroD1 mRNA was widely expressed in PA (82%), with measurable levels found especially in those derived from Pit-1 independent lineages, i.e. corticotroph (5/5) and clinically non-secreting (CNS) adenomas (9/11). According to sq-RT-PCR analysis, overexpression of NeuroD1 compared to normal pituitaries was frequent. Variable nuclear NeuroD1 immunopositivity was also present in about 70% of studied cases. ASH1 mRNA was widely detected in normal pituitaries, in all tumour cell lines and in most PA (84%), with measurable levels in corticotroph (5/5) and CNS (9/11) adenomas, and in a significant subset of PA derived from Pit-1 dependent lineages (9/16). We conclude that: a) NeuroD1 is differentially expressed in PA and its possible ontogenetic and/or pathogenetic implications in non-corticotroph PA are discussed; b) ASH1 is a neuroendocrine marker whose expression is largely conserved in normal and neoplastic pituitary cells.
Collapse
Affiliation(s)
- E Ferretti
- Department of Experimental Medicine, University of L'Aquila, L'Aquila, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Salemi S, Besson A, Eblé A, Gallati S, Pfäffle RW, Mullis PE. New N-terminal located mutation (Q4ter) within the POU1F1-gene (PIT-1) causes recessive combined pituitary hormone deficiency and variable phenotype. Growth Horm IGF Res 2003; 13:264-268. [PMID: 12932747 DOI: 10.1016/s1096-6374(03)00015-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
OBJECTIVE Growth is an inherent property of life. About 10% of the congenital forms of growth retardation and short stature are genetically caused. Beside the gene involved in direct GH-production, there are different candidate genes important for appropriate pituitary development causing combined pituitary hormone deficiency (CPHD). However, severe growth retardation and failure to thrive remain the leading reason for medical assessment in these patients. PATIENTS AND METHODS We report two siblings of a healthy but consanguineous Malaysian family presenting with severe short stature caused by CPHD with a variable phenotype. Importantly, at the beginning the girl presented with isolated GHD, whereas the boy was hypothyroid. As the most common gene alterations responsible for CPHD are within either the PROP-1- or the POU1F1- (PIT-1)-gene these two genes were further studied. RESULTS Subsequent sequencing of the six exons of the POU1F1-gene allowed the identification of a new N-terminal mutation (Q4ter) in these two children. A substitution of C to T induced a change from a glutamine (CAA) to a stop codon (TAA) in exon 1 of the PIT-1 protein. Both affected children were homozygous for the mutation, whereas the mother and father were heterozygous. CONCLUSION We describe two children with autosomal recessive inherited CPHD caused by a new N-terminal located mutation within the PUO1F1-gene. The clinical history of these two children underline the phenotypic variability and support the fact that children with any isolated and/or combined PHD need to be closely followed as at an any time other hormonal deficiencies may occur. In addition, molecular analysis of the possible genes involved might be most helpful for the future follow-up.
Collapse
Affiliation(s)
- Souzan Salemi
- University Children's Hospital, Paediatric Endocrinology, Inselspital, Bern CH-3010, Switzerland
| | | | | | | | | | | |
Collapse
|
42
|
Kawajiri K, Ikuta T, Suzuki T, Kusaka M, Muramatsu M, Fujieda K, Tachibana M, Morohashi KI. Role of the LXXLL-motif and activation function 2 domain in subcellular localization of Dax-1 (dosage-sensitive sex reversal-adrenal hypoplasia congenita critical region on the X chromosome, gene 1). Mol Endocrinol 2003; 17:994-1004. [PMID: 12610109 DOI: 10.1210/me.2002-0360] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Dosage-sensitive sex reversal-adrenal hypoplasia congenita critical region on the X chromosome, gene 1 (Dax-1, NR0B1) is an orphan nuclear receptor that represses transcription by Ad4 binding protein/steroidogenic factor 1 (Ad4BP/SF-1, NR5A1). Observations on human diseases and the phenotypes of mice, in which the corresponding genes have been disrupted, have elucidated essential roles of these two nuclear receptors in differentiation of steroidogenic tissues. However, little is known about how the functions of these factors are regulated. Here we have examined their subcellular localization and have clarified the molecular mechanisms regulating subcellular localization of Dax-1. Prompted by the finding that nuclear localization of Dax-1 correlates with the presence of Ad4BP/SF-1 in the early stages of pituitary development, we have tested the possibility that interaction between the two factors is essential for the nuclear localization of Dax-1. In vitro studies with cultured cells demonstrated that an interaction involving the LXXLL motifs in the N-terminal repeat region of Dax-1 plays a key role in its subcellular localization. In addition, we found that a mutant form of DAX-1 (L466R), from a patient with adrenal hypoplasia congenita, was defective in nuclear localization in spite of having an intact N terminus. Taken together, the results reveal that the subcellular localization of Dax-1 is influenced by the presence of Ad4BP/SF-1, and that two regions of Dax-1 have important roles for this process.
Collapse
Affiliation(s)
- Kaname Kawajiri
- Department of Developmental Biology, National Institute for Basic Biology, Myodaiji-cho, Okazaki 444-8585, Japan
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Keegan CE, Camper SA. Mouse knockout solves endocrine puzzle and promotes new pituitary lineage model. Genes Dev 2003; 17:677-82. [PMID: 12651886 DOI: 10.1101/gad.1085903] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Catherine E Keegan
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI 48109-0638, USA.
| | | |
Collapse
|
44
|
Herzog W, Zeng X, Lele Z, Sonntag C, Ting JW, Chang CY, Hammerschmidt M. Adenohypophysis formation in the zebrafish and its dependence on sonic hedgehog. Dev Biol 2003; 254:36-49. [PMID: 12606280 DOI: 10.1016/s0012-1606(02)00124-0] [Citation(s) in RCA: 134] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Formation of the adenohypophysis in mammalian embryos occurs via an invagination of the oral ectoderm to form Rathke's pouch, which becomes exposed to opposing dorsoventral gradients of signaling proteins governing specification of the different hormone-producing pituitary cell types. One signal promoting pituitary cell proliferation and differentiation to ventral cell types is Sonic hedgehog (Shh) from the oral ectoderm. To study pituitary formation and patterning in zebrafish, we cloned four cDNAs encoding different pituitary hormones, prolactin (prl), proopiomelancortin (pomc), thyroid stimulating hormone (tsh), and growth hormone (gh), and analyzed their expression patterns relative to that of the pituitary marker lim3. prl and pomc start to be expressed at the lateral edges of the lim3 expression domain, before pituitary cells move into the head. This indicates that patterning of the pituitary anlage and terminal differentiation of pituitary cells starts while cells are still organized in a placodal fashion at the anterior edge of the developing brain. Following the expression pattern of prl and pomc during development, we show that no pituitary-specific invagination equivalent to Rathke's pouch formation takes place. Rather, pituitary cells move inwards together with stomodeal cells during oral cavity formation, with medial cells of the placode ending up posterior and lateral cells ending up anterior, resulting in an anterior-posterior, rather than a dorsoventral, patterning of the adenohypophysis. Carrying out loss- and gain-of-function experiments, we show that Shh from the ventral diencephalon plays a crucial role during induction, patterning, and growth of the zebrafish adenohypophysis. The phenotypes are very similar to those obtained upon pituitary-specific inactivation or overexpression of Shh in mouse embryo, suggesting that the role of Shh during pituitary development has been largely conserved between fish and mice, despite the different modes of pituitary formation in the two vertebrate classes.
Collapse
Affiliation(s)
- Wiebke Herzog
- Max-Planck Institute for Immunobiology, Stuebeweg 51, D-79108, Freiburg, Germany
| | | | | | | | | | | | | |
Collapse
|
45
|
Lost and Found: the Pars Intermedia of the Human Pituitary and its Role in the Histogenesis of Silent “Corticotroph” Adenomas. ACTA ACUST UNITED AC 2003. [DOI: 10.1007/978-1-4615-0501-3_13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
46
|
Baumann G. Genetic characterization of growth hormone deficiency and resistance: implications for treatment with recombinant growth hormone. AMERICAN JOURNAL OF PHARMACOGENOMICS : GENOMICS-RELATED RESEARCH IN DRUG DEVELOPMENT AND CLINICAL PRACTICE 2002; 2:93-111. [PMID: 12083945 DOI: 10.2165/00129785-200202020-00003] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Growth failure can be caused by deficient growth hormone production or action. The genes involved in pituitary development, somatotrope function, as well as growth hormone synthesis, secretion, and action have recently been characterized in considerable detail. Familial growth failure has played an important role in identifying these genes, and a large number of mutations adversely affecting the development and function of the growth hormone/insulin-like growth factor axis have been discovered. Inactivating mutations leading to growth retardation in humans have been identified in several pituitary transcription factor genes (HESX1, PITX2, LHX3, PROP1, POU1F1) as well as in genes encoding the growth hormone-releasing hormone receptor (GHRH-R), the G(s) protein alpha subunit (GNAS1), growth hormone itself (GH-1), the growth hormone receptor (GHR), and in a single case each, the insulin-like growth factor I (IGF-I) and the IGF-I receptor. Mutations in pituitary transcription factors cause developmental abnormalities of the pituitary and deficiency of multiple pituitary hormones [growth hormone (GH), prolactin (Prl), thyrotropin (TSH) and lutropin/follitropin (LH/FSH)]. Most of the syndromes respond well to therapy with recombinant GH; exceptions are antibody-mediated resistance in GHD type IA (not all patients) and cases of Laron syndrome (GHR deficiency). Such patients respond to IGF-I therapy. This review summarizes the molecular genetics, functional defects, phenotypes, diagnostic considerations and therapeutic aspects of syndromes associated with mutations in the relevant genes.
Collapse
Affiliation(s)
- Gerhard Baumann
- Center for Endocrinology, Metabolism and Molecular Medicine, Department of Medicine, Northwestern University Medical Schoo1, and Veterans Administration Chicago Health Care System, Chicago, Illinois 60611, USA.
| |
Collapse
|
47
|
Amselem S. Current approaches for deciphering the molecular basis of combined anterior pituitary hormone deficiency in humans. Mol Cell Endocrinol 2002; 197:47-56. [PMID: 12431795 DOI: 10.1016/s0303-7207(02)00278-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
This review focuses on the general strategies currently used to decipher the molecular bases of combined pituitary hormone deficiency (CPHD) of genetic origin. By summarizing illustrative approaches that turned out to be successful for identifying an increasing number of genes involved in CPHD in the human, this article consider predictable obstacles specific to the investigation of these rare and heterogeneous conditions, while underlining the previously unsuspected roles of several of these genes during the development of extrapituitary structures.
Collapse
Affiliation(s)
- Serge Amselem
- Institut National de la Santé et de la Recherche Médicale (Unité 468), Hôpital Henri-Mondor, Créteil 94010, France.
| |
Collapse
|
48
|
Raetzman LT, Ward R, Camper SA. Lhx4 and Prop1 are required for cell survival and expansion of the pituitary primordia. Development 2002; 129:4229-39. [PMID: 12183375 DOI: 10.1242/dev.129.18.4229] [Citation(s) in RCA: 102] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Deficiencies in the homeobox transcription factors LHX4 and PROP1 cause pituitary hormone deficiency in both humans and mice. Lhx4 and Prop1 mutants exhibit severe anterior pituitary hypoplasia resulting from limited differentiation and expansion of most specialized cell types. Little is known about the mechanism through which these genes promote pituitary development. In this study we determined that the hypoplasia in Lhx4 mutants results from increased cell death and that the reduced differentiation is attributable to a temporal shift in Lhx3 activation. In contrast, Prop1 mutants exhibit normal cell proliferation and cell survival but show evidence of defective dorsal-ventral patterning. Molecular genetic analyses reveal that Lhx4 and Prop1 have overlapping functions in early pituitary development. Double mutants exhibit delayed corticotrope specification and complete failure of all other anterior pituitary cell types to differentiate. Thus, Lhx4 and Prop1 have critical, but mechanistically different roles in specification and expansion of specialized anterior pituitary cells.
Collapse
Affiliation(s)
- Lori T Raetzman
- Department of Human Genetics and. Graduate Program in Cell and Molecular Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109-0638, USA
| | | | | |
Collapse
|
49
|
Puebla-Osorio N, Proudman JA, Compton AE, Clements KE, Decuypere E, Vandesande F, Berghman LR. FSH- and LH-cells originate as separate cell populations and at different embryonic stages in the chicken embryo. Gen Comp Endocrinol 2002; 127:242-8. [PMID: 12225765 DOI: 10.1016/s0016-6480(02)00054-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The histological distribution of gonadotrophs containing either LH or FSH, but not both gonadotropins, has been demonstrated before in the juvenile and adult chicken throughout the caudal and cephalic anterior pituitary lobes. In the present investigation, the distribution of FSH- and/or LH-containing gonadotrophs was further investigated in the chicken embryo by use of the same homologous antibodies as used in our earlier study. Fluorescent dual-labeling immunohistochemistry revealed that during embryogenesis LH and FSH reside exclusively in separate gonadotrophs, as has been described before in the post hatch bird. LH-immunoreactive cells were observed for the first time at day 9 of embryogenesis. This is as much as 4 days earlier than the FSH-immunoreactive cells, which appeared at day 13 of embryogenesis. Our results confirm that FSH- and LH-containing gonadotrophs are distributed throughout both lobes of the anterior pituitary. No conspicuous differences were observed between the sexes in any of the aspects investigated. The described situation is unique in that it seems to imply the existence of separate cell lineages for FSH- and LH-producing cells, as opposed to the single gonadotrope lineage described in all other species studied so far, with the exception of bovine. Our data indeed raise the question as to which signaling and/or transcription factors may cause the unique dichotomy observed in the chicken gonadotrophs.
Collapse
Affiliation(s)
- N Puebla-Osorio
- Department of Poultry Science, Texas A&M University, College Station 77843-2472 TX, USA.
| | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Somatotropes and thyrotropes are thought to be derived from the same cellular lineage and the expression of both growth hormone (GH) and thyrotropin (beta TSH) is thought to be dependent upon the same (Pit-1) transcription factor. The presence and comparative distribution of GH- and beta TSH-immunoreactivity in early chick embryos, was therefore investigated, especially as extrapituitary GH-immunoreactive cells are present in some peripheral tissues of early chick embryos prior to the ontogenic differentiation of the pituitary gland. At the end of the first trimester of incubation (embryonic day (ED) 7), GH-immunoreactivity was widespread in the head, particularly in neural tissue. Strong labeling was found in the diencephalon and mesencephalon and in neural ganglia and the trigeminal nerve. beta TSH-immunoreactivity was also present in these tissues, although restricted to the ependymal cells lining the diocoele and mesocoele and absent from mantle layers. It was also present in the cellular layer lining the otic vesicle, which was devoid of GH staining. In contrast, Rathke's pouch, the primordial pituitary gland was without GH- or beta TSH-staining. Control sections incubated with preabsorbed antisera or with pre-immune serum were completely devoid of staining. In the trunk, the epidermal cells were stained for beta TSH, but not for GH. Intense GH-immunoreactivity was present in the ventral and dorsal horns of the spinal cord and was particularly strong in the outer marginal layer. In contrast, beta TSH-immunoreactivity was again restricted to ependymal cells lining the spinal canal, which were devoid of GH-immunoreactivity. Strong GH staining was also present in the dorsal and ventral root ganglia, both of which lacked significant beta TSH staining. In non-neural tissues, both GH and beta TSH staining was present in the crop, although in topographically different cells. beta TSH-immunoreactivity was also present in the cells lining the bronchial ducts and the adluminal linings of the pleural and pericardial cavities. GH-immunoreactivity, in contrast, was absent from the lung but present in the surrounding intracostal muscles and in the Müllerian duct. Both GH- and beta TSH-immunoreactivity was present in liver hepatocytes. These results clearly show, for the first time, the presence of TSH-immunoreactivity in central and peripheral tissues of the ED7 chick embryo, prior to the differentiation of pituitary thyrotropes. They also show that beta TSH- and GH-immunoreactive cells are differentially located within embryonic tissues.
Collapse
Affiliation(s)
- A E Murphy
- Department of Physiology, and Perinatal Research Center, University of Alberta, 7-41 Medical Sciences Building, Edmonton, Alberta T6G 2H7, Canada
| | | |
Collapse
|