66 results on '"Paez-Pereda, Marcelo"'
Search Results
52. Modulation of Growth Hormone and Prolactin Secretion in Trypanosoma cruzi-Infected Mammosomatotrophic Cells
- Author
-
Corrêa-de-Santana, Eliane, primary, Paez-Pereda, Marcelo, additional, Theodoropoulou, Marily, additional, Renner, Ulrich, additional, Stalla, Johanna, additional, Stalla, Günter Karl, additional, and Savino, Wilson, additional
- Published
- 2009
- Full Text
- View/download PDF
53. Interferon-γ inhibits cellular proliferation and ACTH production in corticotroph tumor cells through a novel janus kinases–signal transducer and activator of transcription 1/nuclear factor-kappa B inhibitory signaling pathway
- Author
-
Labeur, Marta, primary, Refojo, Damian, additional, Wölfel, Barbara, additional, Stalla, Johanna, additional, Vargas, Vivian, additional, Theodoropoulou, Marily, additional, Buchfelder, Michael, additional, Paez-Pereda, Marcelo, additional, Arzt, Eduardo, additional, and Stalla, Günter K, additional
- Published
- 2008
- Full Text
- View/download PDF
54. Potential of retinoic acid derivatives for the treatment of corticotroph pituitary adenomas
- Author
-
Labeur, Marta, primary, Paez-Pereda, Marcelo, additional, Arzt, Eduardo, additional, and Stalla, Günter K., additional
- Published
- 2008
- Full Text
- View/download PDF
55. Octreotide, a Somatostatin Analogue, Mediates Its Antiproliferative Action in Pituitary Tumor Cells by Altering Phosphatidylinositol 3-Kinase Signaling and Inducing Zac1 Expression
- Author
-
Theodoropoulou, Marily, primary, Zhang, Jing, additional, Laupheimer, Sandra, additional, Paez-Pereda, Marcelo, additional, Erneux, Christophe, additional, Florio, Tullio, additional, Pagotto, Uberto, additional, and Stalla, Günter K., additional
- Published
- 2006
- Full Text
- View/download PDF
56. Sonic hedgehog regulates CRH signal transduction in the adult pituitary
- Author
-
Vila, Greisa, primary, Papazoglou, Maria, additional, Stalla, Johanna, additional, Theodoropoulou, Marily, additional, Stalla, Günter K., additional, Holsboer, Florian, additional, and Paez-Pereda, Marcelo, additional
- Published
- 2004
- Full Text
- View/download PDF
57. Currently used and investigational drugs for Cushing´s disease
- Author
-
Ciato, Denis, Mumbach, Aizhar G., Paez-Pereda, Marcelo, and Stalla, Günter K.
- Abstract
ABSTRACTIntroduction: Cushing’s disease (CD) is caused by a corticotroph adenoma of the pituitary gland that secretes excess adrenocorticotropic hormone (ACTH) causing increased morbidity and mortality. Surgery is the treatment of choice, but is not always successful. Alternatives include radiotherapy, adrenal surgery, and pharmaceutical therapy. The latter is increasingly gaining momentum due to the recent development of compounds that reduce hypercortisolaemia or its symptoms, acting through different mechanisms.Areas covered: In this article, the authors provide a complete overview of the treatment options for Cushing´s disease, including adrenal-directed, tumor-targeted, and peripheral therapies that are currently used or in development, and discuss their potential advantages and limitations.Expert opinion: Considering the lack of long-term remission in up to half of the patients after surgery, and the delayed response to radiotherapy along with potential side effects, there is a strong need for an effective pharmaceutical treatment. Pasireotide, mifepristone, ketoconazole and metyrapone have been approved by regulatory authorities but their use remains limited due to considerable costs and side effects. Research in this field has focused recently on the improvement of pre-existing drugs and the development of safe new ones. However, few approaches aim at targeting the source of the disease, the ACTH-secreting adenoma.
- Published
- 2017
- Full Text
- View/download PDF
58. Corticotropin-releasing hormone activates ERK1 /2 MAPK in specific brain areas.
- Author
-
Refojo, Damián, Echenique, Carlos, Müller, Marianne B., Reul, Johannes M. H. M., Deussing, Jan M., Wurst, Wolfgang, Sillaber, Inge, Paez-Pereda, Marcelo, Holsboer, Florian, and Arzt, Eduardo
- Subjects
ADRENOCORTICOTROPIC hormone ,PEPTIDE hormones ,PROOPIOMELANOCORTIN ,PROTEIN kinases ,CONFOCAL microscopy ,MITOGENS - Abstract
Corticotropin-releasing hormone (CRH) coordinates hormonal and behavioral responses to stress. The mitogen-activated protein kinase extracellular signal-related kinase ½ (ERK½) mediates several functions in different forebrain structures and recently has been implicated in CRH signaling in cultured cells. To study in vivo CRH-mediated activation of central ERK½, we investigated the expression pattern of the phosphorylated ERK½ (p-ERK½) in the mouse brain after intracerebroventricular CRH injections. As shown by immunohistochemistry and confocal microscopy analysis, CRH administration increased p-ERK½ levels specifically in the CA3 and CA1 hippocampal subfields and basolateral complex of the amygdala, both structures related to external environmental information processing and behavioral aspects of stress. Other regions such as hypothalamic nuclei and the central nucleus of the amygdala, also related to central CRH system but involved in the processing of the ascending visceral information and neuroendocrine-autonomic response to stress, did not show CRH-mediated ERK½ activation. To dissect the involvement of CRH receptor 1 (CRHR1) and CRHR2, we used conditional knockout mice in which Crhr1 is inactivated in the anterior forebrain and limbic structures. The conditional genetic ablation of Crhr1 inhibited the p-ERK½ increase, underlining the involvement of CRHR1 in the CRH-mediated activation. These findings underscore the fact that CRH activates p-ERK½ through CRHR1 only in selected brain regions, pointing to a specific role of this pathway in mediating behavioral adaptation to stress. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
59. Sonic hedgehog regulates CRH signal transduction in the adult pituitary.
- Author
-
Vila, Greisa, Papazoglou, Maria, Stalla, Johanna, Theodoropoulou, Marily, Stalla, Günter K., Holsboer, Florian, and Paez-Pereda, Marcelo
- Subjects
PITUITARY gland ,CORTICOTROPIN releasing hormone ,ADRENOCORTICOTROPIC hormone ,SECRETION ,PITUITARY hormone releasing factors ,HYPOTHALAMO-hypophyseal system - Abstract
Presents a study which investigated the expression and function of sonic hedgehog (Shh) in the adult pituitary gland. Expression of Shh pathway components by corticotroph cells; Stimulation of corticotropin secretion by Shh; Additive effects of Shh and corticotropin-releasing hormone in corticotropin secretion.
- Published
- 2005
- Full Text
- View/download PDF
60. The somatostatin analogue octreotide confers sensitivity to rapamycin treatment on pituitary tumor cells
- Author
-
Marcelo Paez-Pereda, Marily Theodoropoulou, Marco Losa, V. Cerovac, Hadara Rubinfeld, Jose Monteserin-Garcia, Tullio Florio, Michael Buchfelder, Günter K. Stalla, Cerovac, Vesna, Monteserin-Garcia, Jose, Rubinfeld, Hadara, Buchfelder, Michael, Losa, Marco, Florio, Tullio, Paez-Pereda, Marcelo, Stalla, Günter K., and Theodoropoulou, Marily
- Subjects
Adenoma ,medicine.medical_specialty ,Pituitary gland ,Cancer Research ,Octreotide ,Cell Growth Processes ,Pituitary neoplasm ,Biology ,Internal medicine ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Sirolimu ,Pituitary Neoplasms ,Pituitary Neoplasm ,Phosphorylation ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Insulin Receptor Substrate Protein ,Sirolimus ,Antineoplastic Combined Chemotherapy Protocol ,Cell Growth Processe ,Somatostatin receptor ,Pituitary tumors ,Cell Cycle ,Drug Synergism ,medicine.disease ,Up-Regulation ,Oncogene Protein v-akt ,Somatostatin ,Endocrinology ,medicine.anatomical_structure ,Oncology ,Cancer research ,Insulin Receptor Substrate Proteins ,Cyclin-Dependent Kinase Inhibitor p27 ,medicine.drug ,Human - Abstract
Rapamycin and its analogues have significant antiproliferative action against a variety of tumors. However, sensitivity to rapamycin is reduced by Akt activation that results from the ablative effects of rapamycin on a p70 S6K–induced negative feedback loop that blunts phosphoinositide 3-kinase (PI3K)–mediated support for Akt activity. Thus, sensitivity to rapamycin might be increased by imposing an upstream blockade to the PI3K/Akt pathway. Here, we investigated this model using the somatostatin analogue octreotide as a tool to decrease levels of activated Ser473-phosphorylated Akt (pAkt-Ser473) in pituitary tumor cells that express somatostatin receptors. Octreotide increased levels of phosphorylated insulin receptor substrate-1 that were suppressed by rapamycin, subsequently decreasing levels of pAkt-Ser473 through effects on phosphotyrosine phosphatase SHP-1. Octreotide potentiated the antiproliferative effects of rapamycin in immortalized pituitary tumor cells or human nonfunctioning pituitary adenoma cells in primary cell culture, sensitizing tumor cells even to low rapamycin concentrations. Combined treatment of octreotide and rapamycin triggered G1 cell cycle arrest, decreasing E2F transcriptional activity and cyclin E levels by increasing levels of p27/Kip1. These findings show that adjuvant treatment with a somatostatin analogue can sensitize pituitary tumor cells to the antiproliferative effects of rapamycin. Cancer Res; 70(2); 666–74
- Published
- 2010
61. Expression and function of sonic hedgehog pathway components in pituitary adenomas: evidence for a direct role in hormone secretion and cell proliferation
- Author
-
Johanna Stalla, Greisa Vila, Günter K. Stalla, Ulrich Renner, Jörg C. Tonn, Marcelo Paez-Pereda, Marily Theodoropoulou, Marco Losa, Vila, Greisa, Theodoropoulou, Marily, Stalla, Johanna, Tonn, Jörg C., Losa, Marco, Renner, Ulrich, Stalla, Günter K., and Paez-Pereda, Marcelo
- Subjects
Patched ,Adenoma ,Adult ,Male ,Patched Receptors ,medicine.medical_specialty ,Pituitary gland ,animal structures ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Receptors, Cell Surface ,Biology ,Pituitary neoplasm ,Biochemistry ,Patched-2 Receptor ,Endocrinology ,Pituitary adenoma ,Internal medicine ,Cell Line, Tumor ,medicine ,Patched Receptor ,Humans ,Hedgehog Proteins ,Pituitary Neoplasms ,Pituitary Neoplasm ,Sonic hedgehog ,Membrane Protein ,Aged ,Cell Proliferation ,Pituitary tumors ,Biochemistry (medical) ,Membrane Proteins ,Middle Aged ,medicine.disease ,Hormone ,Hedgehog signaling pathway ,Hormones ,Patched-1 Receptor ,medicine.anatomical_structure ,Trans-Activator ,Pituitary Gland ,embryonic structures ,biology.protein ,Trans-Activators ,Female ,Corticotropic cell ,Hedgehog Protein ,Human - Abstract
Context: Sonic hedgehog (Shh) belongs to a family of signaling proteins involved in development and has been recently implicated in cancer. Shh signaling is active in the corticotrophs of the adult pituitary gland, where it cross-talks with the CRH pathway and regulates ACTH secretion. Because developmental pathways are involved in pituitary tumorigenesis, we hypothesized that Shh may be important in pituitary tumors. Objective: The objective of this study was to examine the expression and function of Shh-pathway components in pituitary adenomas. Methods: Using immunohistochemistry, we determined the expression of Shh and its receptors Patched 1 (Ptc1) and Patched 2 (Ptc2) in 55 human pituitary adenomas compared with the normal pituitary gland. The AtT-20 and GH3 pituitary tumor cell lines were used as models for studying the role of Shh on cell proliferation and hormone secretion. The effect of Shh on hormone secretion was confirmed in primary cultures of normal rat pituitaries and human pituitary tumors. Results: Ptc1 and Ptc2 were present, whereas Shh was down-regulated in pituitary adenomas and completely absent in Cushing tumors. Shh inhibited cell proliferation in AtT-20 corticotrophinoma cells and the Shh-specific inhibitor cyclopamine increased proliferation in GH3 mammosomatotrophinoma cells. On the other hand, exogenous administration of Shh increased hormone secretion from normal rat pituitaries, pituitary cell lines, and 10 different pituitary tumors. Conclusions: Our results suggest that Shh might maintain pituitary cells in a nonproliferative state. We conclude that Shh is a newly described hypophysiotropic cytokine and its down-regulation may be involved in the pathogenesis of pituitary adenomas. Copyright © 2005 by The Endocrine Society.
- Published
- 2005
62. A Somatostatin Receptor Subtype-3 (SST 3 ) Peptide Agonist Shows Antitumor Effects in Experimental Models of Nonfunctioning Pituitary Tumors.
- Author
-
Vázquez-Borrego MC, Gupta V, Ibáñez-Costa A, Gahete MD, Venegas-Moreno E, Toledano-Delgado Á, Cano DA, Blanco-Acevedo C, Ortega-Salas R, Japón MA, Barrera-Martín A, Vasiljevic A, Hill J, Zhang S, Halem H, Solivera J, Raverot G, Gálvez MA, Soto-Moreno A, Paez-Pereda M, Culler MD, Castaño JP, and Luque RM
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Animals, Apoptosis drug effects, Cell Proliferation drug effects, Disease Models, Animal, Drug Evaluation, Preclinical, Female, Humans, Janus Kinases metabolism, MAP Kinase Signaling System drug effects, Male, Mice, Mice, Knockout, Middle Aged, Neuroendocrine Tumors metabolism, Neuroendocrine Tumors pathology, Peptides chemistry, Phosphatidylinositol 3-Kinases metabolism, Pituitary Neoplasms metabolism, Pituitary Neoplasms pathology, Signal Transduction, Tumor Cells, Cultured, Young Adult, Neuroendocrine Tumors drug therapy, Peptides pharmacology, Pituitary Neoplasms drug therapy, Receptors, Somatostatin agonists
- Abstract
Purpose: Somatostatin analogues (SSA) are efficacious and safe treatments for a variety of neuroendocrine tumors, especially pituitary neuroendocrine tumors (PitNET). Their therapeutic effects are mainly mediated by somatostatin receptors SST
2 and SST5 . Most SSAs, such as octreotide/lanreotide/pasireotide, are either nonselective or activate mainly SST2 . However, nonfunctioning pituitary tumors (NFPTs), the most common PitNET type, mainly express SST3 and finding peptides that activate this particular somatostatin receptor has been very challenging. Therefore, the main objective of this study was to identify SST3 -agonists and characterize their effects on experimental NFPT models., Experimental Design: Binding to SSTs and cAMP level determinations were used to screen a peptide library and identify SST3 -agonists. Key functional parameters (cell viability/caspase activity/chromogranin-A secretion/mRNA expression/intracellular signaling pathways) were assessed on NFPT primary cell cultures in response to SST3 -agonists. Tumor growth was assessed in a preclinical PitNET mouse model treated with a SST3 -agonist., Results: We successfully identified the first SST3 -agonist peptides. SST3 -agonists lowered cell viability and chromogranin-A secretion, increased apoptosis in vitro , and reduced tumor growth in a preclinical PitNET model. As expected, inhibition of cell viability in response to SST3 -agonists defined two NFPT populations: responsive and unresponsive, wherein responsive NFPTs expressed more SST3 than unresponsive NFPTs and exhibited a profound reduction of MAPK, PI3K-AKT/mTOR, and JAK/STAT signaling pathways upon SST3 -agonist treatments. Concurrently, SSTR3 silencing increased cell viability in a subset of NFPTs., Conclusions: This study demonstrates that SST3 -agonists activate signaling mechanisms that reduce NFPT cell viability and inhibit pituitary tumor growth in experimental models that expresses SST3 , suggesting that targeting this receptor could be an efficacious treatment for NFPTs., (©2019 American Association for Cancer Research.)- Published
- 2020
- Full Text
- View/download PDF
63. Interferon-gamma inhibits cellular proliferation and ACTH production in corticotroph tumor cells through a novel janus kinases-signal transducer and activator of transcription 1/nuclear factor-kappa B inhibitory signaling pathway.
- Author
-
Labeur M, Refojo D, Wölfel B, Stalla J, Vargas V, Theodoropoulou M, Buchfelder M, Paez-Pereda M, Arzt E, and Stalla GK
- Subjects
- Animals, Blotting, Western, Humans, Mice, Pituitary Neoplasms drug therapy, Pro-Opiomelanocortin genetics, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction drug effects, Transcription, Genetic drug effects, Adrenocorticotropic Hormone biosynthesis, Cell Proliferation drug effects, Interferon-gamma pharmacology, Janus Kinases metabolism, NF-kappa B metabolism, Pituitary Neoplasms metabolism, STAT1 Transcription Factor metabolism
- Abstract
Interferon-gamma (IFNG) is a cytokine that exerts potent antiproliferative and tumoricidal effects in a variety of cancers. Moreover, IFNG modulates normal pituitary hormone secretion, and was shown to inhibit the expression of the ACTH precursor POMC in murine ACTH-secreting AtT-2010/21/2008 tumor cells. We have studied the functional role of IFNG on pituitary tumor cells, focusing on the involvement of IFNG in the molecular events leading to the control of POMC transcriptional repression. Herein, it is shown that IFNG inhibits AtT-20 tumor cell proliferation without inducing apoptosis. Unexpectedly, an activated janus kinases-signal transducer and activator of transcription (JAK-STAT1) cascade is required for IFNG inhibitory action on POMC promoter activity. Factor-kappa B (NF-kappaB) is necessary for the inhibitory action of IFNG on Pomc transcription, since loss of NF-kappaB activity with IkappaB super-repressor abolishes this effect. In addition, 1 and 2 IFNG receptor immunoreactivity was detected in human corticotropinoma cells. Interestingly, IFNG inhibits ACTH production from these cells in primary cell culture, without affecting basal ACTH biosynthesis in normal non-tumoral pituitary cells. In conclusion, our data show for the first time that POMC transcription can be negatively regulated by a JAK-STAT1 and NF-kappaB-dependent pathway.
- Published
- 2008
- Full Text
- View/download PDF
64. Signaling processes in tumoral neuroendocrine pituitary cells as potential targets for therapeutic drugs.
- Author
-
Paez-Pereda M, Giacomini D, Echenique C, Stalla GK, Holsboer F, and Arzt E
- Subjects
- Adrenocorticotropic Hormone biosynthesis, Animals, Cytokines metabolism, Drug Delivery Systems, Gene Expression Regulation drug effects, Humans, Prolactin metabolism, Receptors, Cytokine drug effects, Receptors, Cytokine metabolism, Transforming Growth Factor beta metabolism, Neurosecretory Systems drug effects, Neurosecretory Systems physiology, Pituitary Neoplasms drug therapy, Pituitary Neoplasms physiopathology, Signal Transduction drug effects
- Abstract
Pituitary adenomas are neuroendocrine tumors that produce different endocrine and metabolic alterations, including hyperprolactinemia, acromegaly and Cushing's disease. These different clinical features of pituitary tumors are the result of the overproduction of hormones produced by the different pituitary cell types. Recent advances in the understanding of the signaling pathways that control hormone production in pituitary cells provide a source of potential therapeutic targets. In ACTH-secreting cells, the mechanisms that control hormone biosynthesis have been clarified to a great extent, indicating a number of protein kinases and ligand-activated nuclear receptors as targets for experimental drugs. ACTH production requires the activation of signal transduction through the PKA, the MAPK and the CamK pathways. These pathways activate nuclear receptors, including Nur and PPAR gamma. The inhibition of these kinases and nuclear receptors has been shown to produce therapeutic effects in mouse models of Cushing's syndrome. On the other hand, the signaling pathways that control prolactin and growth hormone production also have potential targets. It has been recently shown that SMAD proteins activated by growth factors of the TGF beta and BMP family interact with estrogen receptors to stimulate the proliferation of prolactin and growth hormone-secreting cells. Cytokines that bind to the membrane protein gp130 also stimulate the proliferation of these cells. The inhibition of both of these pathways results in the decrease of tumor growth in animal models of prolactinoma. Therefore, the study of signaling pathways that control hormone production and proliferation is a good source of candidate targets in pituitary tumors.
- Published
- 2005
- Full Text
- View/download PDF
65. Growth factors and cytokines: function and molecular regulation in pituitary adenomas.
- Author
-
Renner U, Paez-Pereda M, Arzt E, and Stalla GK
- Subjects
- Adenoma metabolism, Animals, Humans, Pituitary Neoplasms metabolism, Adenoma physiopathology, Cytokines metabolism, Growth Substances metabolism, Pituitary Neoplasms physiopathology
- Abstract
The growth and functions of the anterior pituitary cells are regulated by hypothalamic factors, peripheral hormones and growth factors. However, the expression of numerous growth factors and cytokines, as well as their receptors, within the anterior pituitary suggests that these factors could be locally involved in the control of pituitary development, function and proliferation by auto-/paracrine mechanisms. In the normal pituitary, intrapituitary factors probably play a role in modifying endocrine signals in the pituitary cells. However, since alterations in growth factor production and receptor expression have been found in pituitary adenomas, these factors may also contribute to the pathophysiology and progression of pituitary tumours. The potential roles of the most important growth factors and cytokines in pituitary adenoma pathogenesis are reviewed.
- Published
- 2004
- Full Text
- View/download PDF
66. Integrating systemic information at the molecular level: cross-talk between steroid receptors and cytokine signaling on different target cells.
- Author
-
Refojo D, Liberman AC, Giacomini D, Carbia Nagashima A, Graciarena M, Echenique C, Paez Pereda M, Stalla G, Holsboer F, and Arzt E
- Subjects
- Animals, Hormones physiology, Humans, Pituitary Neoplasms immunology, Pituitary Neoplasms physiopathology, Prolactinoma immunology, Prolactinoma physiopathology, Signal Transduction physiology, Steroids physiology, Cytokines physiology, Receptor Cross-Talk physiology, Receptors, Steroid physiology
- Abstract
An essential event in immune activation is the increase of cytokines in both plasma and immune tissues. Steroid hormones influence several adaptive responses in both health and disease. Cytokines and steroids have an intimate cross-communication in many systems, making possible a satisfactory adaptive response to environmental changes. The ultimate level of integration of the cytokine-steroids cross-talk is the molecular level. We have demonstrated this in four types of cross-talk mechanisms on different cells in which steroids have major roles: (1) The tumor necrosis factor (TNF)-glucocorticoid receptor (GR) transcriptional interaction in cellular targets of TNF-induced cytotoxicity. TNF potentiates the transactivation activity of GR and the priming with TNF increases the protective action of GR on TNF-induced cytotoxicity. (2) The GR-T cell receptor (TCR) antagonism in GR-TCR-induced T cell apoptosis and its modulation by cAMP. cAMP inhibits the TCR-induced apoptosis through a PKA-CREB-dependent mechanism and potentiates glucocorticoid-induced apoptosis by means of a CREB-independent mechanism. (3) The GR influence on Th1-Th2 cytokine expression and differentiation. Glucocorticoids inhibit the induction of GATA-3 and T-bet transcription factors. (4) The influence of ER/Smad-4 signaling cross-communication on prolactinoma pathogenesis. Physical and functional interactions between Smad-4 and estrogen receptors take place in prolactinoma cells, providing a molecular explanation to link the tumorigenic action of these two important players of prolactinoma pathogenesis. The molecular cross-talk between steroids and transcription factors is the mechanism that provides the basis for the outcome of adaptive responses integrating the systemic information provided by hormones and cytokines.
- Published
- 2003
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.