Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Endocrinology ; 165(8)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38923438

ABSTRACT

The neuroendocrine marker genes Ptprn and Ptprn2 encode protein tyrosine phosphatase receptors N and N2, 2 members of protein tyrosine phosphatase receptors void of enzymatic activity, and whose function and mechanism of action have not been elucidated. To explore the role(s) of Ptprn and Ptprn2 on the hypothalamic-pituitary-adrenal axis, we used mice in which both genes were knocked out (DKO). The focus in this study was on corticotrophs and melanotrophs from the anterior and intermediate lobes of the pituitary gland, respectively. In both sexes, DKO caused an increase in the expression of the corticotroph/melanotroph genes Pomc and Tbx19 and the melanotroph-specific gene Pax7. We also found in vivo and in vitro increased synthesis and release of beta-endorphin, alpha-melanocyte-stimulating hormone, and ACTH in DKO mice, which was associated with increased serum corticosterone levels and adrenal mass. DKO also increased the expression of other melanotroph-specific genes, but not corticotroph-specific genes. The dopaminergic pathway in the hypothalamus and dopaminergic receptors in melanotrophs were not affected in DKO mice. However, hyperplasia of the intermediate lobe was observed in DKO females and males, accompanied by increased proopiomelanocortin immunoreactivity per cell. These results indicate that protein tyrosine phosphatase receptor type N contributes to hypothalamic-pituitary-adrenal function by being involved in processes governing postnatal melanotroph development and Pomc expression.


Subject(s)
Melanotrophs , Mice, Knockout , Pituitary Gland , Pro-Opiomelanocortin , Animals , Mice , Pro-Opiomelanocortin/metabolism , Pro-Opiomelanocortin/genetics , Female , Male , Pituitary Gland/metabolism , Melanotrophs/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Pituitary-Adrenal System/metabolism , Hypothalamo-Hypophyseal System/metabolism , Mice, Inbred C57BL
2.
J Vet Med Sci ; 86(1): 71-76, 2024 Jan 10.
Article in English | MEDLINE | ID: mdl-37981318

ABSTRACT

A 22-year and 9-month-old female Grevy's zebra (Equus grevyi) showed signs of polyuria, polydipsia, glucosuria, and muscle atrophy. Blood tests revealed hyperglycemia, hypertriglyceridemia, electrolyte imbalance, high levels of adrenocorticotropic hormone (ACTH) and cortisol, and low levels of hormones secreted by the pituitary pars distalis. Pathological examinations revealed a pituitary gland tumor and bilateral adrenal cortical hyperplasia. Pituitary tumor cells showed immunoreactivity for α-melanocyte-stimulating hormone and ACTH. The deposition of amyloid ß was observed in the parenchyma and vascular walls of the cerebrum. The zebra showed clinical signs of pituitary pars intermedia dysfunction and was histopathologically diagnosed with pituitary gland melanotroph adenoma. This case report provides insight into neoplastic and endocrine diseases associated with the aging of a zebra.


Subject(s)
Adenoma , Pituitary Neoplasms , Female , Animals , Pituitary Neoplasms/veterinary , Melanotrophs/metabolism , Melanotrophs/pathology , Amyloid beta-Peptides , Equidae , Pituitary Gland/metabolism , Adrenocorticotropic Hormone/metabolism , Adenoma/veterinary , Adenoma/pathology
3.
Nat Commun ; 12(1): 2028, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33795686

ABSTRACT

Germline mutations in BRAF and other components of the MAPK pathway are associated with the congenital syndromes collectively known as RASopathies. Here, we report the association of Septo-Optic Dysplasia (SOD) including hypopituitarism and Cardio-Facio-Cutaneous (CFC) syndrome in patients harbouring mutations in BRAF. Phosphoproteomic analyses demonstrate that these genetic variants are gain-of-function mutations leading to activation of the MAPK pathway. Activation of the MAPK pathway by conditional expression of the BrafV600E/+ allele, or the knock-in BrafQ241R/+ allele (corresponding to the most frequent human CFC-causing mutation, BRAF p.Q257R), leads to abnormal cell lineage determination and terminal differentiation of hormone-producing cells, causing hypopituitarism. Expression of the BrafV600E/+ allele in embryonic pituitary progenitors leads to an increased expression of cell cycle inhibitors, cell growth arrest and apoptosis, but not tumour formation. Our findings show a critical role of BRAF in hypothalamo-pituitary-axis development both in mouse and human and implicate mutations found in RASopathies as a cause of endocrine deficiencies in humans.


Subject(s)
Gain of Function Mutation , Hypopituitarism/genetics , Hypothalamus/metabolism , Pituitary Gland/metabolism , Proto-Oncogene Proteins B-raf/genetics , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , Child , Child, Preschool , Corticotrophs/cytology , Corticotrophs/metabolism , Ectodermal Dysplasia/genetics , Facies , Failure to Thrive/genetics , HEK293 Cells , Heart Defects, Congenital/genetics , Humans , Infant , MAP Kinase Signaling System/genetics , Melanotrophs/cytology , Melanotrophs/metabolism , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins B-raf/metabolism , Exome Sequencing/methods
4.
Thyroid ; 31(5): 850-858, 2021 05.
Article in English | MEDLINE | ID: mdl-33191870

ABSTRACT

Background: Thyrotropin (TSH) is well known as the hormone of the anterior pituitary thyrotrophs responsible for acting in the thyroid gland, where it stimulates synthesis and release of thyroid hormones through Gs and Gq/11 protein coupled TSH receptors (TSHRs). Methods: In this study, we examined whether the functional TSHRs are also expressed in cultured rat pituitary cells, using double immunocytochemistry, quantitative reverse transcription-polymerase chain reaction analysis, cAMP and hormone measurements, and single-cell calcium imaging. Results: Double immunocytochemistry revealed the expression of TSHRs in cultured corticotrophs and melanotrophs, in addition to previously identified receptors in folliculostellate cells. The functional coupling of these receptors to the Gq/11 signaling pathway was not observed, as demonstrated by the lack of TSH activation of IP3-dependent calcium mobilization in these cells when bathed in calcium-deficient medium. However, TSH increased cAMP production in a time- and concentration-dependent manner and facilitated calcium influx in single corticotrophs and melanotrophs, indicating their coupling to the Gs signaling pathway. Consistent with these findings, TSH stimulated adrenocorticotropin and ß-endorphin release in male and female pituitary cells in a time- and concentration-dependent manner without affecting the expression of proopiomelanocortin gene. Conclusions: These results indicate that TSH is a potential paracrine modulator of anterior pituitary corticotrophs and melanotrophs, controlling the exocytotic but not the transcriptional pathway in a cAMP/calcium influx-dependent manner.


Subject(s)
Corticotrophs/metabolism , Melanotrophs/metabolism , Pro-Opiomelanocortin/genetics , Receptors, Thyrotropin/genetics , Thyrotrophs/metabolism , Animals , Cells, Cultured , Immunohistochemistry , Paracrine Communication , Pituitary Gland, Anterior/metabolism , Pro-Opiomelanocortin/metabolism , Rats , Receptors, Thyrotropin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Single-Cell Analysis
5.
Cell Calcium ; 79: 11-19, 2019 05.
Article in English | MEDLINE | ID: mdl-30772686

ABSTRACT

Rat melanotrophs express several types of voltage-gated and ligand-gated calcium channels, although mechanisms involved in the maintenance of the resting intracellular Ca2+ concentration ([Ca2+]i) remain unknown. We analyzed mechanisms regulating resting [Ca2+]i in dissociated rat melanotrophs by Ca2+-imaging and patch-clamp techniques. Treatment with antagonists of L-type, but not N- or P/Q-type voltage-gated Ca2+ channels (VGCCs) as well as removal of extracellular Ca2+ resulted in a rapid and reversible decrease in [Ca2+]i, indicating constitutive Ca2+ influx through L-type VGCCs. Reduction of extracellular Na+ concentration (replacement with NMDG+) similarly decreased resting [Ca2+]i. When cells were champed at -80 mV, decrease in the extracellular Na+ resulted in a positive shift of the holding current. In cell-attached voltage-clamp and whole-cell current-clamp configurations, the reduction of extracellular Na+ caused hyperpolarisation. The holding current shifted in negative direction when extracellular K+ concentration was increased from 5 mM to 50 mM in the presence of K+ channel blockers, Ba2+ and TEA, indicating cation nature of persistent conductance. RT-PCR analyses of pars intermedia tissues detected mRNAs of TRPV1, TRPV4, TRPC6, and TRPM3-5. The TRPV channel blocker, ruthenium red, shifted the holding current in positive direction, and significantly decreased the resting [Ca2+]i. These results indicate operation of a constitutive cation conductance sensitive to ruthenium red, which regulates resting membrane potential and [Ca2+]i in rat melanotrophs.


Subject(s)
Calcium Channels, L-Type/metabolism , Calcium/metabolism , Melanotrophs/metabolism , Sodium/metabolism , Animals , Male , Patch-Clamp Techniques , Rats , Rats, Wistar , Ruthenium Red/pharmacology , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism
6.
Development ; 143(13): 2376-88, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27226320

ABSTRACT

Sox2 mutations are associated with pituitary hormone deficiencies and the protein is required for pituitary progenitor proliferation, but its function has not been well characterized in this context. SOX2 is known to activate expression of Six6, encoding a homeodomain transcription factor, in the ventral diencephalon. Here, we find that the same relationship likely exists in the pituitary. Moreover, because Six6 deletion is associated with a similar phenotype as described here for loss of Sox2, Six6 appears to be an essential downstream target of SOX2 in the gland. We also uncover a second role for SOX2. Whereas cell differentiation is reduced in Sox2 mutants, some endocrine cells are generated, such as POMC-positive cells in the intermediate lobe. However, loss of SOX2 here results in complete downregulation of the melanotroph pioneer factor PAX7, and subsequently a switch of identity from melanotrophs to ectopic corticotrophs. Rescuing proliferation by ablating the cell cycle negative regulator p27 (also known as Cdkn1b) in Sox2 mutants does not restore melanotroph emergence. Therefore, SOX2 has two independent roles during pituitary morphogenesis; firstly, promotion of progenitor proliferation, and subsequently, acquisition of melanotroph identity.


Subject(s)
Cell Lineage , Pituitary Gland/cytology , Pituitary Gland/embryology , SOXB1 Transcription Factors/metabolism , Stem Cells/cytology , Animals , Cell Count , Cell Proliferation , Corticotrophs/cytology , Corticotrophs/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Down-Regulation/genetics , Gene Deletion , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Melanotrophs/cytology , Melanotrophs/metabolism , Mice, Inbred C57BL , Models, Biological , Morphogenesis/genetics , PAX7 Transcription Factor/metabolism , Pro-Opiomelanocortin/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Stem Cells/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
7.
Endocrinology ; 155(9): 3538-49, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24926820

ABSTRACT

Cushing's disease (CD) is a rare condition in which hypercortisolemia is secondary to excessive ACTH release from a pituitary corticotroph adenoma. CD is associated with significant morbidity and mortality, and a safe therapy that effectively targets the pituitary tumor is still lacking. Retinoic acid (RA) and dopamine agonists (DAs) have recently been considered as monotherapy in CD patients, and satisfactory results have been reported, albeit in a limited number of patients. Given the permissive role of RA on the dopamine receptor type-2 (DRD2), the aim of the present study was to see whether a combination of 9-cis RA and the DA bromocriptine (Br) might represent a possible treatment for CD. Here we show that 9-cis RA induces a functional DRD2 in the pituitary corticotroph cell line AtT20, and increases cell sensitivity to Br via a mechanism only partially related to corticotroph-to-melanotroph transdifferentiation. In addition, 9-cis RA and Br act synergistically to modulate cell viability, with favorable implications for clinical use. In nearly 45% of corticotropinoma-derived primary cultures, the combined administration of 9-cis RA and Br lowered the steady-state level of the ACTH precursor proopiomelanocortin (POMC) more efficiently than either of the drugs alone. In conclusion, the effects of a combination of 9-cis RA and Br on ACTH synthesis/secretion and cell viability in AtT20, and on POMC transcriptional activity in human corticotropinomas might represent a suitable starting point for assessing the potential of this treatment regimen for ACTH-secreting pituitary adenomas. This study thus has potentially important implications for novel therapeutic approaches to CD.


Subject(s)
Adrenocorticotropic Hormone/metabolism , Cell Proliferation , Cell Transdifferentiation , Corticotrophs/cytology , Melanotrophs/cytology , Pituitary ACTH Hypersecretion/physiopathology , Receptors, Dopamine D2/genetics , Tretinoin/metabolism , Adolescent , Adult , Animals , Corticotrophs/metabolism , Humans , Male , Melanotrophs/metabolism , Mice , Middle Aged , Models, Biological , Pituitary ACTH Hypersecretion/genetics , Pituitary ACTH Hypersecretion/metabolism , Promoter Regions, Genetic , Receptors, Dopamine D2/metabolism , Up-Regulation , Young Adult
8.
Ann Anat ; 195(6): 512-21, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23938266

ABSTRACT

Cetaceans share peculiar features of their pituitary glands, with a complete separation of pars distalis and pars nervosa by a dural septum and the absence of an intermediate lobe and cleft. In most mammals the pars intermedia is the main source of circulating α-melanocyte stimulating hormone (α-MSH), derived from a large precursor called proopiomelanocortin (POMC), which also generates adrenocorticotropic hormone (ACTH) in the adenohypophysis. The lack of an intermediate lobe in cetaceans led us to investigate whether their glands are able to produce α-MSH, and if this hormone is secreted by a distinct population of melanotrophs or by corticotrophs in the pars distalis. Immunofluorescence evidences seem to support the first assumption, with ACTH-immunoreactive (-ir) elements rarely overlapping with α-MSH-ir ones. The discovery of a population of true melanotrophs in the hypophysis of some odontocetes underscores the need for further research on the melanocortin system of cetaceans.


Subject(s)
Dolphins/anatomy & histology , Melanotrophs/ultrastructure , Pituitary Gland, Posterior/ultrastructure , Adrenocorticotropic Hormone/metabolism , Animals , Bottle-Nosed Dolphin/physiology , Cell Count , Common Dolphins/physiology , Corticotrophs/metabolism , Dolphins/physiology , Fluorescent Antibody Technique , Immunoenzyme Techniques , Immunohistochemistry , Melanotrophs/metabolism , Pituitary Gland, Posterior/cytology , Pituitary Gland, Posterior/metabolism , Pro-Opiomelanocortin/metabolism , Sheep , Species Specificity , Swine , Tissue Fixation , alpha-MSH/metabolism
9.
Mol Endocrinol ; 27(7): 1103-12, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23754801

ABSTRACT

The identification of a stable pool of progenitor/stem cells in the adult pituitary has renewed the interest of identifying mechanisms for maintenance of pituitary cells throughout life. Whereas developmental studies have shown that progenitor expansion is the major source of new differentiated cells during pituitary organogenesis, the contribution of these progenitors for maintenance of the adult tissue is not clear although progenitors were clearly involved in cell expansion following end-organ ablation, notably after adrenalectomy and/or gonadectomy. We have used a genetic trick that eliminates dividing cells by apoptosis in order to assess the contribution of differentiated corticotropes and melanotropes for maintenance of their population in the adult pituitary. The system relies on chromosome instability created by the action of the Cre recombinase on inverted loxP sites. Expression of Cre recombinase in corticotropes and melanotropes led to progressive loss of corticotropes whereas melanotropes were unaffected. Because the Cre transgene is not expressed in progenitors, the data indicate that maintenance of the adult corticotrope pool is primarily due to self-duplication of differentiated cells. In contrast, melanotropes do not divide. Maintenance of corticotropes by self-duplication contrasts with the reported proliferative response of undifferentiated cells observed after adrenalectomy. If corticotrope reentry into cell cycle constitutes a normal mechanism to maintain the adult corticotrope pool, this same mechanism may also be perturbed during corticotrope adenoma development in Cushing's disease.


Subject(s)
Aging/physiology , Cell Lineage , Pituitary Gland/cytology , Adrenalectomy , Animals , Cell Differentiation , Cell Division , Cell Proliferation , Corticotrophs/cytology , Corticotrophs/metabolism , Integrases , Melanotrophs/cytology , Melanotrophs/metabolism , Mice , Models, Biological , Pro-Opiomelanocortin/metabolism , Stem Cells/cytology , Stem Cells/metabolism
10.
Mol Cell Endocrinol ; 372(1-2): 49-56, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23541636

ABSTRACT

Prolonged exposure to stress elicits profound effects on homeostasis that may lead to cryptogenic disorders such as chronic fatigue syndrome. To investigate the pathophysiology associated with the syndrome, we used a rat continuous stress (CS) model where the pituitary represents one of the most affected organs. Here we found that mRNA for VGF (non-acronymic), a member of the granin family, was induced specifically in the intermediate lobe (IL). This was matched by a concomitant increase at the peptide/protein level assessed by C-terminal antibody. Furthermore, the up-regulation of VGF was confirmed by immunohistochemistry in a subset of melanotrophs. VGF expression was altered in the IL of rats receivingthe dopamine D2 receptor agonist bromocriptine or the antagonist sulpiride. In vitro, dopamine dose-dependently decreased the mRNA levels in cultured melanotrophs. These findings suggest that VGF expression under CS is negatively regulated by dopaminergic neurons projecting from the hypothalamus.


Subject(s)
Dopamine/physiology , Melanotrophs/metabolism , Neuropeptides/genetics , Stress, Physiological , Transcriptional Activation , Animals , Bromocriptine/pharmacology , Cells, Cultured , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Dopaminergic Neurons/metabolism , Gene Expression , Hypothalamus/cytology , Hypothalamus/metabolism , Male , Neuropeptides/metabolism , Pituitary Gland, Intermediate/cytology , Pituitary Gland, Intermediate/drug effects , Pituitary Gland, Intermediate/metabolism , Primary Cell Culture , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Sulpiride/pharmacology
11.
Gen Comp Endocrinol ; 177(3): 315-21, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22248443

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is, despite its name, also found outside the central nervous system (CNS), but the functional significance of this observation is largely unknown. This review concerns the expression of BDNF in the pituitary gland. While the presence of the neurotrophin in the mammalian pituitary gland is well documented its functional significance remains obscure. Studies on the pars intermedia of the pituitary of the amphibian Xenopus laevis have shown that BDNF is produced by the neuroendocrine melanotrope cells, its expression is physiologically regulated, and the melanotrope cells themselves express receptors for the neurotrophin. The neurotrophin has been shown to act as an autocrine factor on the melanotrope to promote cell growth and regulate gene expression. In doing so BDNF supports the physiological function of the cell to produce and release α-melanophore-stimulating hormone for the purpose of adjusting the animal's skin color to that of its background.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Melanotrophs/cytology , Melanotrophs/metabolism , Xenopus laevis/metabolism , Animals , Gene Expression
12.
Biomed Res ; 32(3): 225-35, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21673453

ABSTRACT

Dopamine regulates the synthesis and secretion of prolactin and α-MSH/ß-endorphin in lactotrophs and melanotrophs, respectively. While a predominant dopamine receptor, D2R, is known to be expressed in both the anterior and intermediate lobes of the pituitary gland, no previous immunohistochemical studies have shown the existence of D2R in the plasma membrane of pituitary endocrine cells. The present study clearly demonstrated a selective localization of the D2R immunoreactivity in primary cilia of lactotrophs and melanotrophs in the mouse adenohypophysis. Another immunoreactivity of D2R was found along the plasma membrane of melanotrophs. The intensity of immunoreactivity for D2R in the primary cilia of lactrotrophs changed during the estrous cycle and with genital conditions in contrast to a consistent immunolabeling in the melanotrophs. Since there is accumulating evidence that the primary cilium functions as a sensory device at a cellular level, the D2R-expressing primary cilia in the pituitary gland may be involved in the sensation of dopamine and dopaminergic compounds-though their involvement differs between the anterior and intermediate lobes.


Subject(s)
Cell Membrane/ultrastructure , Immunohistochemistry/methods , Lactotrophs/ultrastructure , Melanotrophs/ultrastructure , Receptors, Dopamine D2/ultrastructure , Animals , Cell Membrane/metabolism , Cilia/metabolism , Cilia/ultrastructure , Female , In Situ Hybridization , Lactotrophs/metabolism , Male , Melanotrophs/metabolism , Mice , Microscopy, Electron, Scanning , Pregnancy , RNA, Messenger/analysis , Receptors, Dopamine D2/metabolism , Tyrosine 3-Monooxygenase/metabolism
13.
Endocrinology ; 152(6): 2321-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21447633

ABSTRACT

We tested whether double cortin-like kinase-short (DCLK-short), a microtubule-associated Ser/Thr kinase predominantly expressed in the brain, is downstream of the ERK signaling pathway and is involved in proopiomelanocortin gene (POMC) expression in endocrine pituitary melanotrope cells of Xenopus laevis. Melanotropes form a well-established model to study physiological aspects of neuroendocrine plasticity. The amphibian X. laevis adapts its skin color to the background light intensity by the release of α-MSH from the melanotrope cell. In frogs on a white background, melanotropes are inactive but they are activated during adaptation to a black background. Our results show that melanotrope activation is associated with an increase in DCLK-short mRNA and with phosphorylation of DCLK-short at serine at position 30 (Ser-30). Upon cell activation phosphorylated Ser-30-DCLK-short was translocated from the cytoplasm into the nucleus, and the ERK blocker U0126 inhibited this process. The mutation of Ser-30 to alanine also inhibited the translocation and reduced POMC expression, whereas overexpression stimulated POMC expression. This is the first demonstration of DCLK-short in a native endocrine cell. We conclude that DCLK-short is physiologically regulated at both the level of its gene expression and protein phosphorylation and that the kinase is effectively regulating POMC gene expression upon its ERK-mediated phosphorylation.


Subject(s)
Cell Nucleus/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Melanotrophs/metabolism , Pro-Opiomelanocortin/genetics , Protein Serine-Threonine Kinases/metabolism , Up-Regulation , Xenopus Proteins/metabolism , Xenopus laevis/metabolism , Animals , Cell Nucleus/genetics , Cells, Cultured , Phosphorylation , Pro-Opiomelanocortin/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Transport , Xenopus Proteins/genetics , Xenopus laevis/genetics
14.
Mol Endocrinol ; 25(2): 348-59, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21193556

ABSTRACT

Cell-specific expression of the pituitary proopiomelanocortin (POMC) gene depends on the combination of tissue- and cell-restricted transcription factors such as Pitx1 and Tpit. These factors act on the proximal POMC promoter together with transcription factors that integrate inputs from signaling pathways. We now report the identification of an upstream enhancer in the POMC locus that is targeted by the same subset of transcription factors, except Pitx1. This enhancer located at -7 kb in the mouse POMC gene is highly dependent on Tpit for activity. Whereas Tpit requires Pitx1 for action on the promoter, it acts on the -7-kb enhancer as homodimers binding to a palindromic Tpit response element (TpitRE). Both half-sites of the TpitRE palindrome and Tpit homodimerization are required for activity. In vivo, the enhancer exhibits preferential activity in corticotrope cells of the anterior lobe whereas the promoter exhibits preference for intermediate lobe melanotropes. The enhancer is conserved among different species with the TpitRE palindrome localized at the center of conserved sequences. However, the mouse and human -7-kb enhancers do not exhibit conservation of hormone responsiveness and may differ in their relative importance for POMC expression. In summary, pituitary expression of the POMC gene relies on an upstream enhancer that complements the activity of the proximal promoter with Tpit as the major regulator of both regulatory regions.


Subject(s)
Corticotrophs/metabolism , Enhancer Elements, Genetic , Homeodomain Proteins/metabolism , Pro-Opiomelanocortin/genetics , Response Elements , T-Box Domain Proteins/metabolism , Animals , COS Cells , Chlorocebus aethiops , Gene Expression , Homeodomain Proteins/genetics , Humans , Melanotrophs/metabolism , Mice , Mice, Transgenic , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Pituitary Gland/metabolism , Polymerase Chain Reaction , Pro-Opiomelanocortin/metabolism , Promoter Regions, Genetic , Rats , Signal Transduction , T-Box Domain Proteins/genetics
15.
Gen Comp Endocrinol ; 170(1): 57-67, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20888821

ABSTRACT

This review gives an overview of the functioning of the hypothalamo-hypophyseal neuroendocrine interface in the pituitary neurointermediate lobe, as it relates to melanotrope cell function in two amphibian species, Rana ridibunda and Xenopus laevis. It primarily but not exclusively concerns the work of two collaborating laboratories, the Laboratory for Molecular and Cellular Neuroendocrinology (University of Rouen, France) and the Department of Cellular Animal Physiology (Radboud University Nijmegen, The Netherlands). In the course of this review it will become apparent that Rana and Xenopus have, for the most part, developed the same or similar strategies to regulate the release of α-melanophore-stimulating hormone (α-MSH). The review concludes by highlighting the molecular and cellular mechanisms utilized by thyrotropin-releasing hormone (TRH) to activate Rana melanotrope cells and the function of autocrine brain-derived neurotrophic factor (BDNF) in the regulation of Xenopus melanotrope cell function.


Subject(s)
Melanocyte-Stimulating Hormones/metabolism , Melanotrophs/cytology , Melanotrophs/metabolism , Neuroendocrine Cells/metabolism , Animals , Neuroendocrine Cells/cytology , Pro-Opiomelanocortin/metabolism , Rana ridibunda , Xenopus laevis
16.
Gen Comp Endocrinol ; 170(1): 156-61, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20888824

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is expressed in the mammalian pituitary gland, in both the anterior and intermediate lobes, where its functional significance is unknown. Melanotrope cells in the intermediate pituitary lobe of the amphibian Xenopus laevis also produce BDNF, which co-exists in secretory granules with α-melanophore-stimulating hormone (α-MSH), a peptide that causes pigment dispersion in dermal melanophores during adaptation of the toad to a dark background. Xenopus melanotropes are highly plastic, undergoing very strong growth to support the high biosynthesis and release of α-MSH in black-adapted animals. In this study we have tested our hypothesis that this enhanced growth of the melanotrope is maintained by autocrine release of BDNF. Furthermore, since the extracellular-regulated kinase (ERK) pathway is a major component of BDNF signaling in neuronal plasticity, we investigated its involvement in melanotrope cell growth. For these purposes melanotropes were treated for 3 days in vitro, with either an anti-BDNF serum or a recombinant tropomyosin-receptor kinase B (TrkB) receptor fragment to eliminate released BDNF, or with the ERK inhibitor U0126. We also applied a novel inhibitor of the TrkB receptor, cyclotraxin-B, to test this receptor's involvement in melanotrope cell growth regulation. All treatments markedly reduced melanotrope cell growth. Therefore, we conclude that autocrine release of BDNF and subsequent TrkB-dependent ERK-mediated signaling is important for melanotrope cell growth during its physiologically induced activation.


Subject(s)
Brain-Derived Neurotrophic Factor/chemistry , Brain-Derived Neurotrophic Factor/metabolism , Melanotrophs/metabolism , Amino Acid Sequence , Animals , Brain-Derived Neurotrophic Factor/immunology , Butadienes/pharmacology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Humans , Immune Sera/immunology , Immune Sera/pharmacology , Melanotrophs/drug effects , Molecular Sequence Data , Nitriles/pharmacology , Peptides, Cyclic/pharmacology , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Kinases/pharmacology , Sequence Homology, Amino Acid , Signal Transduction/drug effects , Xenopus laevis
17.
Biochimie ; 93(3): 528-32, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21118709

ABSTRACT

The p24 proteins function in early secretory pathway transport processes, but their exact role is unclear. In physiologically activated Xenopus melanotrope cells, a representative of each p24 subfamily (p24α(3), -ß(1), -γ(3), -δ(2)) is upregulated coordinately with the major melanotrope cargo, proopiomelanocortin (POMC), whereas two other p24s (p24γ(2) and -δ(1)) are also expressed, but not coordinately with POMC. Using melanotrope-specific transgene expression, we here find that the roles of both p24γ(2) and p24δ(1) in the transport, glycosylation, sulphation and cleavage of POMC are different from those of their upregulated subfamily relatives (p24γ(3) and p24δ(2), respectively). Thus, even p24 proteins from the same subfamily have distinct functions in secretory cargo biosynthesis.


Subject(s)
Vesicular Transport Proteins/classification , Vesicular Transport Proteins/metabolism , Xenopus Proteins/classification , Xenopus Proteins/metabolism , Animals , Melanotrophs/metabolism , Pro-Opiomelanocortin/biosynthesis , Pro-Opiomelanocortin/metabolism , Protein Processing, Post-Translational , Transgenes/genetics , Vesicular Transport Proteins/genetics , Xenopus Proteins/genetics , Xenopus laevis
18.
Mol Endocrinol ; 25(1): 117-27, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21084383

ABSTRACT

The pituitary gland contains six distinct hormone-secreting cell types that are essential for basic physiological processes including fertility and responding to stress. Formation of hormone-secreting cells during development relies on Notch signaling to prevent progenitors from prematurely differentiating. The nature of the signal curtailing Notch signaling in the pituitary is unknown, but a good candidate is the endocytic adaptor protein NUMB. NUMB targets Notch for proteolytic degradation, but it also has a broad range of actions, including stabilizing adherens junctions through interactions with cadherins and influencing cell proliferation by stabilizing expression of the tumor suppressor protein p53. Here, we show that NUMB and its closely related homolog, NUMBLIKE, are expressed in undifferentiated cells during development and later in gonadotropes in the anterior lobe and melanotropes of the intermediate lobe. All four isoforms of NUMB, are detectable in the pituitary, with the shorter forms becoming more prominent after adolescence. Conditionally deleting Numb and Numblike in the intermediate lobe melanotropes with Pomc Cre mice dramatically alters the morphology of cells in the intermediate lobe, coincident with impaired localization of adherens junctions proteins including E-CADHERIN, N-CADHERIN, ß-CATENIN, and α-CATENIN. Strikingly, the border between posterior and intermediate lobes is also disrupted. These mice also have disorganized progenitor cells, marked by SOX2, but proliferation is unaffected. Unexpectedly, Notch activity appears normal in conditional knockout mice. Thus, Numb is critical for maintaining cell-cell interactions in the pituitary intermediate lobe that are essential for proper cell placement.


Subject(s)
Cell Movement , Gene Deletion , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Pituitary Gland, Intermediate/cytology , Pituitary Gland, Intermediate/embryology , Pro-Opiomelanocortin/metabolism , Stem Cells/cytology , Animals , Cell Adhesion , Cell Proliferation , Gene Expression Regulation, Developmental , Gonadotrophs/cytology , Gonadotrophs/metabolism , Integrases/metabolism , Intracellular Signaling Peptides and Proteins , Melanotrophs/cytology , Melanotrophs/metabolism , Membrane Proteins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Pituitary Gland, Intermediate/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Notch/metabolism , Stem Cells/metabolism
19.
J Neuroendocrinol ; 23(3): 261-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21129045

ABSTRACT

The extracellular signal-regulated kinase (ERK) pathway is important in the regulation of neuronal plasticity, although a role for the kinase in regulating plasticity of neuroendocrine systems has not been examined. The melanotroph cells in the pars intermedia of pituitary gland of the amphibian Xenopus laevis are highly plastic, undergoing very strong growth to support the high biosynthetic and secretory activity involving α-melanophore-stimulating hormone (α-MSH), a peptide that causes pigment dispersion in dermal melanophores during the adaptation of the animal to a dark background. In the present study, we tested our hypothesis that ERK-signalling is involved in the regulation of melanotroph cell function during black-background adaptation, namely in the production of pro-opiomelanocortin (POMC), the precursor of α-MSH. Using western blot analyses, we found elevated levels of the activated (phosphorylated) form of ERK in melanotrophs of black- versus white-adapted animals. Treatment of melanotrophs in vitro with the mitogen-activated protein kinase kinase inhibitor U0126 markedly reduced ERK phosphorylation and lowered the transcription as well as the translation of POMC. This same treatment also reduced the expression of BDNF transcript IV and of the immediate early genes c-Fos and Nur77. We conclude that ERK-mediated signalling is important for the maintenance of the melanotroph cells in an active state.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/physiology , Melanotrophs/metabolism , Pro-Opiomelanocortin/biosynthesis , Adaptation, Biological/genetics , Adaptation, Biological/physiology , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Butadienes/pharmacology , Cells, Cultured , Color , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Nitriles/pharmacology , Phosphorylation/drug effects , Pituitary Gland/metabolism , Pituitary Gland, Intermediate/drug effects , Pituitary Gland, Intermediate/metabolism , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Protein Kinase Inhibitors/pharmacology , Tissue Distribution , Xenopus laevis
20.
Gen Comp Endocrinol ; 169(2): 123-9, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20736010

ABSTRACT

Pituitary melanotrope cells of the amphibian Xenopus laevis are neuroendocrine cells regulating the animal's skin color adaptation through secretion of α-melanophore-stimulating hormone (α-MSH). To fulfill this function optimally, the melanotrope cell undergoes plastic changes in structure and secretory activity in response to changed background light conditions. Xenopus melanotrope cells display Ca(2+) oscillations that are thought to drive α-MSH secretion and gene expression. They also produce brain-derived neurotrophic factor (BDNF), which stimulates in an autocrine way the biosynthesis of the α-MSH precursor, pro-opiomelanocortin (POMC). We have used this physiological adaptation mechanism as a model to investigate the role of BDNF in the regulation of Ca(2+) kinetics and Ca(2+)-dependent gene expression. By dynamic video imaging of isolated cultured melanotropes we demonstrated that BDNF caused a dose-dependent increase in Ca(2+) oscillation frequency up to 64.7±2.3% of control level. BDNF also induced a transient Ca(2+) peak in Ca(2+)-free medium, which was absent when calcium stores were blocked by thapsigargin and 2-aminoethoxydiphenyl borate, indicating that BDNF stimulates acute release of Ca(2+) from IP(3)-sensitive intracellular Ca(2+) stores. Moreover, we show that thapsigargin inhibits the expression of BDNF transcript IV (by 61.1±28.8%) but does not affect POMC transcript. We conclude that BDNF mobilizes Ca(2+) from IP(3)-sensitive intracellular Ca(2+) stores and propose the possibility that the resulting Ca(2+) oscillations selectively stimulate expression of the BDNF gene.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Calcium/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Neuroendocrine Cells/drug effects , Neuroendocrine Cells/metabolism , Animals , Cells, Cultured , Melanotrophs/cytology , Melanotrophs/drug effects , Melanotrophs/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...