Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
2.
Pituitary ; 26(1): 73-93, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36422846

ABSTRACT

CONTEXT: Pituitary tumors are the third most common brain tumor and yet there is no standardization of the surveillance schedule and assessment modalities after transsphenoidal surgery. EVIDENCE ACQUISITION: OVID, EMBASE and the Cochrane Library databases were systematically screened from database inception to March 5, 2020. Inclusion and exclusion criteria were designed to capture studies examining detection of pituitary adenoma recurrence in patients 18 years of age and older following surgical resection with curative intent. EVIDENCE SYNTHESIS: A total of 7936 abstracts were screened, with 812 articles reviewed in full text and 77 meeting inclusion criteria for data extraction. A pooled analysis demonstrated recurrence rates at 1 year, 5 years and 10 years for non-functioning pituitary adenomas (NFPA; N = 3533 participants) were 1%, 17%, and 33%, for prolactin-secreting adenomas (PSPA; N = 1295) were 6%, 21%, and 28%, and for growth-hormone pituitary adenomas (GHPA; N = 1257) were 3%, 8% and 13%, respectively. Rates of recurrence prior to 1 year were 0% for NFPA, 1-2% for PSPA and 0% for GHPA. The mean time to disease recurrence for NFPA, PSPA and GHPA were 4.25, 2.52 and 4.18 years, respectively. CONCLUSIONS: This comprehensive review of the literature quantified the recurrence rates for commonly observed pituitary adenomas after transsphenoidal surgical resection with curative intent. Our findings suggest that surveillance within 1 year may be of low yield. Further clinical trials and cohort studies investigating cost-effectiveness of surveillance schedules and impact on quality of life of patients under surveillance will provide further insight to optimize follow-up.


Subject(s)
Adenoma , Lactotrophs , Pituitary Neoplasms , Somatotrophs , Humans , Adolescent , Adult , Pituitary Neoplasms/surgery , Pituitary Neoplasms/pathology , Lactotrophs/pathology , Somatotrophs/pathology , Quality of Life , Neoplasm Recurrence, Local/epidemiology , Adenoma/surgery , Adenoma/pathology , Retrospective Studies
3.
Front Endocrinol (Lausanne) ; 13: 955100, 2022.
Article in English | MEDLINE | ID: mdl-35983516

ABSTRACT

Objective: A 22-year-old man complaining erectile dysfunction underwent transsphenoidal surgery for a 2.7 cm sellar mass with total resection and was confirmed at pathology to have a lactotroph pituitary neuroendocrine tumor (PiNET). Postoperatively, the patient's PRL remained at high level and therefore accepted high-dose dopamine receptor agonist (DA) therapy. After over 3 months of bromocriptine (BRC) (15mg/day) and over 3 years of cabergoline (CAB) (3mg/week) therapy, the patient's prolactin (PRL) never achieved long-term normalization. He was diagnosed with DA-resistant lactotroph PitNET. Method: In this study, the patient was given hydroxychloroquine (HCQ) (200 mg/d) and CAB (3 mg/w) in combination for four months. His PRL level was tested by blood test every month. Results: Taking the combination therapy of HCQ and CAB, the patient's uncontrolled PRL level was normalized within one month and was maintained at the normal level thereafter. Pituitary magnetic resonance imaging (MRI) images with enhancement showed no recurrence. The patient also regained normal sexual function. Discussion: This is the first report on the combination of HCQ with CAB for the effective treatment of DA-resistant lactotroph pituitary neuroendocrine tumor in a patient, which might provide a novel treatment strategy for clinical management.


Subject(s)
Lactotrophs , Neuroendocrine Tumors , Pituitary Neoplasms , Prolactinoma , Adult , Cabergoline/therapeutic use , Ergolines/therapeutic use , Humans , Hydroxychloroquine/therapeutic use , Lactotrophs/pathology , Male , Neuroendocrine Tumors/complications , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/surgery , Pituitary Neoplasms/complications , Pituitary Neoplasms/drug therapy , Pituitary Neoplasms/surgery , Prolactin , Prolactinoma/complications , Prolactinoma/drug therapy , Prolactinoma/pathology , Young Adult
4.
Clin Neuropathol ; 41(5): 211-218, 2022.
Article in English | MEDLINE | ID: mdl-35575415

ABSTRACT

This case report concerns a 31-year-old male with an aggressive pituitary tumor who presented initially with bitemporal hemianopsia and slightly elevated prolactin. On magnetic resonance imaging of the brain, there was a sellar mass with parasellar invasion to the lateral aspects of the internal carotid arteries, compressing the optic chiasm. On histopathological analysis, the diagnosis was made of a densely granulated lactotroph pituitary tumor with a Ki67 proliferation rate of 15%, a mitotic count of 6/10 high-power fields, and p53 positivity. Based on these features, the tumor was classified as a grade 2b tumor according to the Trouillas classification, and a more aggressive behavior of the tumor could be expected. In order to anticipate a future need for alternative drug treatments, the following analyses were undertaken: MGMT methylation (present) as well as the expression of estrogen receptor (negative), programmed-death ligand 1 (60 - 70% positive tumor cells), vascular endothelial growth factor-A and somatostatin receptor 2 (both positive). There was regrowth of residual tumor tissue, and the treatment consisted thus far of repeat surgery, cabergoline, pasireotide, and radiotherapy. Chemotherapy with temozolomide could not yet be initiated due to a concurrent infertility treatment. This case is unique because the tumor displays atypical characteristics, both in terms of morphology and behavior. It also illustrates how pathologists can play an important role in determining the diagnosis, prognosis, and possibilities for targeted therapy.


Subject(s)
Lactotrophs , Pituitary Neoplasms , Adult , Cabergoline/therapeutic use , Humans , Ki-67 Antigen , Lactotrophs/pathology , Male , Pituitary Neoplasms/pathology , Prolactin/therapeutic use , Receptors, Estrogen/therapeutic use , Tumor Suppressor Protein p53/therapeutic use , Vascular Endothelial Growth Factor A/therapeutic use
5.
Endocr Relat Cancer ; 29(6): 359-373, 2022 05 11.
Article in English | MEDLINE | ID: mdl-35324456

ABSTRACT

Among pituitary adenomas, prolactinomas are the most frequently diagnosed (about 50%). Dopamine agonists are generally effective in the treatment of prolactinomas. However, a subset of about 25% of patients does not respond to these agents. The management of drug-resistant prolactinomas remains a challenge for endocrinologists and new inhibitory treatments are needed. Pituitary activins inhibit lactotroph function. Its expression and action were found reduced in animal models of lactotroph hyperplasia (female mice overexpressing the B subunit of the human chorionic gonadotrophin and female mice knockout for dopamine receptor type 2). In these models, an oophorectomy avoids prolactinoma development. Hormonal replacement with oestradiol and/or progesterone is not enough to reach the tumor size observed in transgenic females. We postulated that the loss of gonadal inhibins after an oophorectomy contributes to prevent hyperplasia development. Here, we demonstrated that an oophorectomy at 2 months age recovers the following in adulthood: (i) pituitary activin expression, (ii) activin receptor expression specifically in lactotroph population, (iii) activin biological activity in lactotrophs with a concomitant reduction of Pit-1 expression. To summarize, when an oophorectomy is performed, inhibins are lost and the inhibitory action of pituitary activins on lactotroph population is recovered, helping to prevent lactotroph hyperplasia development. These results emphasize the importance of the inhibitory action of activins on lactotroph function, positioning activins as a good therapeutic target for the treatment of resistant prolactinomas.


Subject(s)
Lactotrophs , Pituitary Neoplasms , Prolactinoma , Activins/metabolism , Adult , Animals , Female , Humans , Hyperplasia , Inhibins/metabolism , Inhibins/therapeutic use , Lactotrophs/metabolism , Lactotrophs/pathology , Mice , Ovariectomy , Pituitary Neoplasms/metabolism , Prolactin/metabolism , Prolactinoma/metabolism , Prolactinoma/prevention & control
7.
PLoS One ; 13(9): e0204727, 2018.
Article in English | MEDLINE | ID: mdl-30261014

ABSTRACT

Pituitary adenoma is a common intracranial neoplasm that is observed in approximately 10% of unselected individuals at autopsy. Prolactin-producing adenomas, i.e., prolactinomas, comprise approximately 50% of all pituitary adenomas and represent the most common class of pituitary tumor. Multiple observations suggest that estrogens may contribute to development of prolactinoma; however, direct evidence for a causal role of estrogens in prolactinoma etiology is lacking. Rat models of estrogen-induced prolactinoma have been utilized extensively to identify the factors, pathways and processes that are involved in pituitary tumor development. The objective of this study was to localize to high resolution Ept7 (Estrogen-induced pituitary tumor), a quantitative trait locus (QTL) that controls lactotroph responsiveness to estrogens and was mapped to rat chromosome 7 (RNO7) in an intercross between BN and ACI rats. Data presented and discussed herein localize the Ept7 causal variant(s) to a 1.91 Mb interval of RNO7 that contains two protein coding genes, A1bg and Myc, and Pvt1, which yields multiple non-protein coding transcripts of unknown function. The Ept7 orthologous region in humans is located at 8q24.21 and has been linked in genome wide association studies to risk of 8 distinct epithelial cancers, including breast, ovarian, and endometrial cancers; 3 distinct types of B cell lymphoma; multiple inflammatory and autoimmune diseases; and orofacial cleft defects. In addition, the Ept7 locus in humans has been associated with variation in normal hematologic and development phenotypes, including height. Functional characterization of Ept7 should ultimately enhance our understanding of the genetic etiology of prolactinoma and these other diseases.


Subject(s)
Adenoma , Chromosomes, Mammalian/genetics , Estrogens , Lactotrophs/metabolism , Pituitary Neoplasms , Quantitative Trait Loci , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Animals , Estrogens/genetics , Estrogens/metabolism , Female , Genome-Wide Association Study , Humans , Hyperplasia , Lactotrophs/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Pituitary Neoplasms/genetics , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Rats
8.
World Neurosurg ; 120: e241-e246, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30138730

ABSTRACT

BACKGROUND: Tumor recurrence or incomplete resection in nonfunctioning pituitary adenomas (NFPAs) is relatively common. However, predictive factors of tumor recurrence in NFPAs are not well established. We evaluated possible factors related to tumor recurrence in a large cohort of NFPAs at a single pituitary neurosurgery center. METHODS: A retrospective analysis was conducted of 410 medical records of patients with NFPAs treated by transsphenoidal surgery between 2000 and 2014. RESULTS: Among the participants, 210 were female (51.0%). A total of 14.1% had giant adenomas. Null-cell pituitary adenomas (n = 239; 58.9%) were the most frequent, followed by silent gonadotroph adenomas (n = 112; 27.3%). Null-cell adenomas were more frequent in women (P = 0.008) and silent gonadotroph adenomas were more frequent in men (P = 0.004). Recurrence was not related to sex or age. Tumor recurrence occurred more often among silent corticotropic adenomas and giant adenomas (hazard ratio 2.45; P < 0.0001 and hazard ratio 2.35; P = 0.001, respectively). Silent thyrotrophic adenoma presented a comparable frequency of recurrence of silent corticotropic adenomas, despite having borderline significance (P = 0.07). CONCLUSIONS: NFPA tumors have a high heterogeneous hormonal profile and may have prognostic importance. Silent corticotropic adenomas and giant adenomas present a high rate of recurrence.


Subject(s)
Adenoma/surgery , Neoplasm Recurrence, Local/epidemiology , Neurosurgical Procedures , Pituitary Neoplasms/surgery , Adenoma/metabolism , Adenoma/pathology , Adrenocorticotropic Hormone/metabolism , Adult , Corticotrophs/metabolism , Corticotrophs/pathology , Female , Follicle Stimulating Hormone/metabolism , Gonadotrophs/metabolism , Gonadotrophs/pathology , Human Growth Hormone/metabolism , Humans , Immunohistochemistry , Lactotrophs/metabolism , Lactotrophs/pathology , Luteinizing Hormone/metabolism , Male , Middle Aged , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Prognosis , Proportional Hazards Models , Retrospective Studies , Sex Factors , Somatotrophs/metabolism , Somatotrophs/pathology , Thyrotrophs/metabolism , Thyrotrophs/pathology , Thyrotropin/metabolism , Tumor Burden
9.
Mol Cell Endocrinol ; 456: 51-61, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-28089822

ABSTRACT

Since the presence of microRNAs was first observed in normal pituitary, the majority of scientific publications addressing their role and the function of microRNAs in the pituitary have been based on pituitary tumor studies. In this review, we briefly describe the involvement of microRNAs in the synthesis of pituitary hormones and we present a comprehensive inventory of microRNA suppressors and inducers of pituitary tumors. Finally, we summarize the functional role of microRNAs in tumorigenesis, progression and aggressiveness of pituitary tumors, mechanisms contributing to the regulation (transcription factors, genomic modifications or epigenetic) or modulation (pharmacological treatment) of microRNAs in these tumors, and the interest of thoroughly studying the expression of miRNAs in body fluids.


Subject(s)
Carcinogenesis/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Pituitary Gland/metabolism , Pituitary Hormones/genetics , Pituitary Neoplasms/genetics , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Humans , Lactotrophs/metabolism , Lactotrophs/pathology , MicroRNAs/metabolism , Mutation , Pituitary Gland/physiopathology , Pituitary Hormones/metabolism , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/physiopathology , Ribonuclease III/genetics , Ribonuclease III/metabolism , Signal Transduction , Somatotrophs/metabolism , Somatotrophs/pathology , Thyrotrophs/metabolism , Thyrotrophs/pathology
10.
PLoS One ; 10(10): e0140699, 2015.
Article in English | MEDLINE | ID: mdl-26509893

ABSTRACT

Recent evidence indicated that alcohol exposure during the fetal period increases the susceptibility to tumor development in mammary and prostate tissues. Whether fetal alcohol exposure increases the susceptibility to prolactin-producing tumor (prolactinoma) development in the pituitary was studied by employing the animal model of estradiol-induced prolactinomas in Fischer 344 female rats. We employed an animal model of fetal alcohol exposure that simulates binge alcohol drinking during the first two trimesters of human pregnancy and involves feeding pregnant rats with a liquid diet containing 6.7% alcohol during gestational day 7 to day 21. Control rats were pair-fed with isocaloric liquid diet or fed ad libitum with rat chow diet. Adult alcohol exposed and control female offspring rats were used in this study on the day of estrus or after estrogen treatment. Results show that fetal alcohol-exposed rats had increased levels of pituitary weight, pituitary prolactin (PRL) protein and mRNA, and plasma PRL. However, these rats show decreased pituitary levels of dopamine D2 receptor (D2R) mRNA and protein and increased pituitary levels of D2R promoter methylation. Also, they show elevated pituitary mRNA levels of DNA methylating genes (DNMT1, DNMT3b, MeCP2) and histone modifying genes (HDAC2, HDAC4, G9a). When fetal alcohol exposed rats were treated neonatally with a DNA methylation inhibitor 5-Aza deoxycytidine and/or a HDAC inhibitor trichostatin-A their pituitary D2R mRNA, pituitary weights and plasma PRL levels were normalized. These data suggest that fetal alcohol exposure programs the pituitary to increase the susceptibility to the development of prolactinomas possibly by enhancing the methylation of the D2R gene promoter and repressing the synthesis and control of D2R on PRL-producing cells.


Subject(s)
Epigenesis, Genetic , Pituitary Gland/pathology , Prenatal Exposure Delayed Effects/blood , Prolactin/biosynthesis , Receptors, Dopamine D2/metabolism , Animals , Cell Proliferation , CpG Islands/genetics , DNA Methylation/genetics , Estrogens/pharmacology , Female , Histones/metabolism , Lactotrophs/drug effects , Lactotrophs/metabolism , Lactotrophs/pathology , Mitogens/pharmacology , Organ Size , Pituitary Gland/metabolism , Pregnancy , Prolactin/blood , Promoter Regions, Genetic , Protein Processing, Post-Translational , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Inbred F344 , Receptors, Dopamine D2/genetics , Transcription, Genetic
11.
Adv Exp Med Biol ; 846: 37-59, 2015.
Article in English | MEDLINE | ID: mdl-25472533

ABSTRACT

Dysregulation of the signaling pathways that govern lactotrope biology contributes to tumorigenesis of prolactin (PRL)-secreting adenomas, or prolactinomas, leading to a state of pathological hyperprolactinemia. Prolactinomas cause hypogonadism, infertility, osteoporosis, and tumor mass effects, and are the most common type of neuroendocrine tumor. In this review, we highlight signaling pathways involved in lactotrope development, homeostasis, and physiology of pregnancy, as well as implications for signaling pathways in pathophysiology of prolactinoma. We also review mutations found in human prolactinoma and briefly discuss animal models that are useful in studying pituitary adenoma, many of which emphasize the fact that alterations in signaling pathways are common in prolactinomas. Although individual mutations have been proposed as possible driving forces for prolactinoma tumorigenesis in humans, no single mutation has been clinically identified as a causative factor for the majority of prolactinomas. A better understanding of lactotrope-specific responses to intracellular signaling pathways is needed to explain the mechanism of tumorigenesis in prolactinoma.


Subject(s)
Carcinogenesis , Lactotrophs/pathology , Lactotrophs/physiology , Animals , Carcinogenesis/genetics , Cell Differentiation/genetics , Female , Homeostasis/genetics , Humans , Pituitary Neoplasms/genetics , Pregnancy , Prolactinoma/genetics , Signal Transduction
12.
Mol Cell Endocrinol ; 383(1-2): 193-202, 2014 Mar 05.
Article in English | MEDLINE | ID: mdl-24373949

ABSTRACT

In the pituitary the activation of cyclic adenosine 3'-5'-monophosphate (cAMP) dependent pathways generates proliferative signals in somatotrophs, whereas in pituitary cells of other lineages its effect remains uncertain. Moreover, the specific role of the two main cAMP effectors, protein kinase A (PKA) and exchange proteins directly activated by cAMP (Epac), has not been defined. Aim of this study was to investigate the effect of cAMP on pituitary adenomatous cells proliferation and to identify PKA and Epac differential involvement. We found that cAMP increased DNA synthesis and cyclin D1 expression in somatotropinomas, whereas it reduced both parameters in prolactinomas and nonfunctioning adenomas, these effects being replicated in corresponding cell lines. Moreover, the divergent cAMP effects were mimicked by Epac and PKA analogs, which activated Rap1 and CREB, respectively. In conclusion, we demonstrated that cAMP exerted opposite effects on different pituitary cell types proliferation, these effects being mediated by both Epac and PKA.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP/metabolism , Gene Expression Regulation, Neoplastic , Guanine Nucleotide Exchange Factors/genetics , Pituitary Gland/metabolism , Protein Subunits/genetics , Adenoma/genetics , Adenoma/metabolism , Adenoma/pathology , Animals , Cell Proliferation , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclin D1/genetics , Cyclin D1/metabolism , Gonadotrophs/metabolism , Gonadotrophs/pathology , Guanine Nucleotide Exchange Factors/metabolism , Humans , Lactotrophs/metabolism , Lactotrophs/pathology , Pituitary Gland/pathology , Pituitary Neoplasms/genetics , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Prolactinoma/genetics , Prolactinoma/metabolism , Prolactinoma/pathology , Protein Subunits/metabolism , Rats , Signal Transduction , Somatotrophs/metabolism , Somatotrophs/pathology , rap1 GTP-Binding Proteins/genetics , rap1 GTP-Binding Proteins/metabolism
13.
Mol Cell Endocrinol ; 349(2): 105-10, 2012 Feb 26.
Article in English | MEDLINE | ID: mdl-22056414

ABSTRACT

The existence of a functional bone morphogenetic protein (BMP) system in the pituitary has been recognized. Recent studies have provided evidence that BMPs elicit differential actions in the regulation of prolactin (PRL) and adrenocorticotropin (ACTH) release in lactotropinoma and corticotropinoma cells, respectively. BMPs play a key role in the modulation of somatostatin receptor (SSTR) sensitivity of lactosomatotrope cells in an autocrine/paracrine manner. In addition, SSTR action enhances BMP responsiveness in corticotrope cells. The functional link between BMP receptor signaling and SSTR actions may be crucial for individual tolerance to somatostatin analogs for controlling PRL and ACTH production. Adjustment of the endogenous SSTR sensitivity may be an effective strategy to inhibit the growth activity and hormonal productivity of intractable pituitary tumors.


Subject(s)
Adenoma/metabolism , Bone Morphogenetic Proteins/physiology , Corticotrophs/metabolism , Lactotrophs/metabolism , Pituitary Neoplasms/metabolism , Adenoma/drug therapy , Adenoma/pathology , Adrenocorticotropic Hormone/metabolism , Animals , Autocrine Communication , Corticotrophs/drug effects , Corticotrophs/pathology , Humans , Lactotrophs/drug effects , Lactotrophs/pathology , Paracrine Communication , Pituitary Neoplasms/drug therapy , Pituitary Neoplasms/pathology , Prolactin/metabolism , Receptors, Somatostatin/metabolism , Signal Transduction , Somatostatin/analogs & derivatives , Somatostatin/metabolism , Somatostatin/pharmacology
14.
Endocr Pathol ; 22(3): 150-4, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21584660

ABSTRACT

Neuroendocrine secretory protein-55 (NESP-55) is a recently described member of the chromogranin family and appears to be a marker of the constitutive secretory pathway in certain neural, neuroendocrine, and endocrine cell types. It has been shown to be selectively expressed in tumors differentiating towards the adrenal chromaffin and pancreatic islet cell phenotypes. The highest levels of NESP-55 expression, at least in animals, appear to be in the adrenal medulla and the pituitary gland. However, very little is known about the status of NESP-55 expression in pituitary adenomas. We therefore studied the immunohistochemical profile of NESP-55 expression in a series of 30 well-characterized pituitary adenomas (five each of FSH/LH and ACTH, four GH, three TSH, seven prolactin, and six null cells). All tumors were positive for one or more generic marker(s) (chromogranin A, synaptophysin, neuron-specific enolase) of neuroendocrine differentiation. All pituitary adenomas selected for study were stained for NESP-55 with appropriate positive and negative controls. NESP-55 immunoreactivity, seen as brown finely granular cytoplasmic staining of the tumor cells with prominent perinuclear accentuation, was graded as focal (<10% tumor cells staining), moderate (10-50% tumor cells staining), and diffuse (>50% tumor cell staining). Four of seven prolactinomas were positive for NESP-55 (one focal, two moderate, and one diffuse). Two of four GH adenomas were also positive (one focal and one diffuse) while only 1/5 FSH tumors showed a moderately intense immunoreactivity. All other pituitary adenomas were completely negative for NESP-55. Our results indicate that, in human pituitary adenomas, NESP-55 has a more restricted pattern of expression than that of chromogranins A and B. Since immunohistochemical expression of NESP-55 is largely confined to prolactinomas and GH adenomas, it raises the possibility that NESP-55 may somehow be involved in the secretory pathways of these specific cell types.


Subject(s)
Adenoma/metabolism , GTP-Binding Protein alpha Subunits, Gs/metabolism , Immunohistochemistry , Pituitary Neoplasms/metabolism , Adenoma/chemistry , Adenoma/pathology , Adrenocorticotropic Hormone/metabolism , Chromogranins/metabolism , GTP-Binding Protein alpha Subunits, Gs/analysis , Humans , Lactotrophs/metabolism , Lactotrophs/pathology , Organ Specificity , Pituitary Neoplasms/chemistry , Pituitary Neoplasms/pathology , Somatotrophs/metabolism , Somatotrophs/pathology , Tissue Distribution
15.
Mol Endocrinol ; 25(1): 92-103, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21106881

ABSTRACT

Dopamine agonist resistance or intolerance is encountered in approximately 20% of prolactinoma patients. Because human epidermal growth factor receptor 2 (HER2)/ErbB2 is overexpressed in prolactinomas and ErbB receptor ligands regulate prolactin (PRL) gene expression, we tested the role of HER2/ErbB2 in prolactinoma hormone regulation and adenoma cell proliferation to assess the rationale for targeting this receptor for prolactinoma therapy. As we showed prolactinoma HER2 overexpression, we generated constitutively active HER2-stable GH3 cell transfectants (HER2CA). PRL mRNA levels were induced approximately 250-fold and PRL secretion was enhanced 100-fold in HER2CA cells, which also exhibited increased proliferation. Lapatinib, a dual tyrosine kinase inhibitor (TKI) of both epidermal growth factor receptor (EGFR)/ErbB1 and HER2, blocked receptor signaling, and suppressed PRL expression more than gefitinib, a TKI of EGFR/ErbB1. Lapatinib also suppressed colony formation in soft agar more than gefitinib. Oral lapatinib treatment caused tumor shrinkage and serum PRL suppression both in HER2CA transfectant-inoculated Wistar-Furth rats and in estrogen-induced Fischer344 rat prolactinomas. In cultured human cells derived from resected prolactinoma tissue, lapatinib suppressed both PRL mRNA expression and secretion. These results demonstrate that prolactinoma HER2 potently induces PRL and regulates experimental prolactinoma cell proliferation. Because pituitary HER2 signaling is abrogated by TKIs, this receptor could be an effective target for prolactinoma therapy.


Subject(s)
Molecular Targeted Therapy/methods , Prolactinoma/metabolism , Prolactinoma/therapy , Receptor, ErbB-2/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gefitinib , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lactotrophs/drug effects , Lactotrophs/metabolism , Lactotrophs/pathology , Lapatinib , Prolactin/genetics , Prolactin/metabolism , Prolactinoma/genetics , Quinazolines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Signal Transduction/drug effects
16.
Environ Toxicol ; 26(4): 359-63, 2011 Aug.
Article in English | MEDLINE | ID: mdl-20082443

ABSTRACT

Freshwater fish Heteropneustes fossilis (H. fossilis) were subjected to 5.76 µg/L (80% of 96 h LC(50) ) and 1.44 µg/L (20% of 96 h LC(50) ) of cypermethrin for short-term (96 h) and long-term (28 days) duration, respectively. Plasma calcium of H. fossilis exposed for short term (96 h) to cypermethrin exhibited no change at 24 h. The levels indicate a decrease in plasma calcium at 48 h. This response persists till the close of experiment (96 h). No change has been noticed throughout the experiment in the histological structure and nuclear volume of prolactin cells of short-term cypermethrin treated fish. Long-term exposure of cypermethrin to fish provoked hypocalcemia. The prolactin cells remain unchanged till 7 days following cypermethrin treatment. After 14 days, the nuclear volume exhibits an increase and the cells exhibit degranulation. These changes increase progressively 21 days onwards. Also, few degenerating cells are discerned after 28 days.


Subject(s)
Catfishes/physiology , Insecticides/toxicity , Lactotrophs/drug effects , Prolactin/blood , Pyrethrins/toxicity , Animals , Calcium/blood , Dose-Response Relationship, Drug , Lactotrophs/metabolism , Lactotrophs/pathology , Lethal Dose 50 , Water Pollutants, Chemical/toxicity
17.
Pituitary ; 13(3): 256-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20405324

ABSTRACT

Rosai-Dorfman disease is a rare, histiocytic proliferative disorder of unknown etiology commonly affecting lymph nodes. Extranodal lesions with or without nodal involvement also occur. We report the case of a 63 year-old woman with disseminated Rosai-Dorfman disease involving the neurohypophysis and associated with adenohypophysial PRL cell hyperplasia.


Subject(s)
Histiocytosis, Sinus/diagnosis , Pituitary Gland, Posterior/pathology , Female , Histiocytosis, Sinus/pathology , Humans , Hyperplasia/pathology , Lactotrophs/metabolism , Lactotrophs/pathology , Middle Aged
18.
J Endocrinol Invest ; 33(5): 325-31, 2010 May.
Article in English | MEDLINE | ID: mdl-19955848

ABSTRACT

BACKGROUND: The term double pituitary adenomas (DPA) is usually referred to those rare lesions showing two distinct cellular components. Genetic background may sustain the proliferation of more than one cell at the same time but no information is available on the presence of aip mutations in these patients. AIM: We report the prevalence and the endocrinological, neuroradiological, histopathological and genetic features of DPA detected in a large surgical series. The contribution of pituitary transcription factor immunostains in DPA was also evaluated. SUBJECTS AND METHODS: One-hundred-forty-four patients undergoing surgery for tumors of the sellar region were evaluated. Histopathology, immunohistochemistry and the mutational analysis for the entire coding region of the AIP and MEN1 genes were performed. RESULTS: One-hundred-seventeen patients out of 144 had a pituitary adenoma. DPA was found in 3 (2.6%) out of 117 patients with pituitary adenoma. Immunohistochemistry and transcription factors analysis demonstrated two not yet described histotype associations in DPA. The coexistence of somatotroph-lactotroph and silent mammosomatotroph histotype in 1 case and the coexistence of sparsely granulated lactotroph and null cell adenomas in the remaining two cases were first identified. Sequencing data for the coding region of the aip and the menin gene resulted in wild type sequences in all patients with DPA. CONCLUSIONS: The prevalence of DPA observed in our unselected surgical series is not negligible (2.6%). Furthermore, the evaluation of the treatment outcome would suggest that the clinical management of DPAs requires a careful diagnostic approach and follow- up.


Subject(s)
Adenoma/epidemiology , Pituitary Neoplasms/epidemiology , Adaptor Proteins, Signal Transducing , Adenoma/genetics , Adenoma/surgery , Adult , Carrier Proteins/biosynthesis , Carrier Proteins/genetics , DNA Mutational Analysis , DNA, Neoplasm/genetics , Guanylate Kinases , Humans , Immunohistochemistry , Lactotrophs/pathology , Magnetic Resonance Imaging , Male , Middle Aged , Pituitary Neoplasms/genetics , Pituitary Neoplasms/surgery , Prolactinoma/genetics , Prolactinoma/pathology , Prolactinoma/surgery , Proto-Oncogene Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Treatment Outcome
19.
Endocrinology ; 150(9): 4443-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19477936

ABSTRACT

Thyroid hormone is important for pituitary development and maintenance. We previously reported that in the Pax8(-/-) mouse model of congenital hypothyroidism, lactotrophs are almost undetectable, whereas the thyrotrophs exhibit hyperplasia and hypertrophy. Because the latter might be caused by an overstimulation of thyrotrophs with TRH, we analyzed TRH-R1(-/-)Pax8(-/-) double-knockout mice, which miss a functional thyroid gland and the TRH transducing receptor-1 at pituitary target sites. Interestingly, in these double mutants, the hypertrophy and hyperplasia of the thyrotrophs still persist, suggesting that the phenotype is rather a direct consequence of the athyroidism of the animals. The increased expression of TSH in the Pax8(-/-) mice was paralleled by a strongly up-regulated expression of deiodinase type 2 (Dio2) in thyrotrophic cells. Moreover, coexpression of TSH and Dio2 could also be demonstrated in the pituitary of wild-type mice, underlining the important role of this enzyme in the negative feedback regulation of TSH by thyroid hormone. As another consequence of the athyroidism in the mutant mice, tyrosine hydroxylase mRNA expression was found to be also highly up-regulated in thyrotrophic cells of the pituitaries from Pax8(-/-) mice, whereas the transcript levels in the hypothalamus were not affected. Accordingly, tyrosine hydroxylase protein levels, enzyme activities, and ultimately dopamine concentrations were found to be strongly increased in the pituitaries of Pax8(-/-) mice compared with wild-type animals. These findings may explain in part the reduced number of lactotrophs found in the pituitary of athyroid Pax8(-/-) mice and suggest a novel paracrine regulatory mechanism of lactotroph activity.


Subject(s)
Pituitary Gland/cytology , Pituitary Hormones/metabolism , Thyrotrophs/metabolism , Animals , Congenital Hypothyroidism/metabolism , Dopamine/metabolism , Lactotrophs/pathology , Male , Mice , Mice, Knockout , PAX8 Transcription Factor , Paired Box Transcription Factors/deficiency , Pituitary Gland/pathology , Thyrotrophs/pathology , Tyrosine 3-Monooxygenase/metabolism
20.
Colloids Surf B Biointerfaces ; 71(2): 187-93, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19286358

ABSTRACT

Laser capture microdissection (LCM) technology combined with immunohistochemistry (immuno-LCM) is a valuable tool to obtain specific target cell populations and therefore this technique enables more accurate proteomic profile. In this study, we optimized the regular immuno-LCM technique to isolate and stain pure prolactin cells from either normal human pituitary (n=6) or prolactioma (n=11). Compared with the routine procedure, more intense and specific staining could be obtained when sections were pretreated with 0.2% Triton X-100 for 4 min. Interestingly, longer pretreatment (0.2% Triton X-100 for 10 min) or higher concentration (2% Triton X-100 for 4 and 10 min) greatly impaired labeling intensity and cell shape. Further scanning electron microscope study revealed that the component extracted from the cell surface by Triton X-100 was lipid. Using the optimized immuno-LCM technique, more pure prolactin cells could be isolated and prepared for further proteomic analysis. Taken together, we reported an optimized immuno-LCM technique that could effectively dissect pure target cells in different type pituitary adenomas for further proteomics analysis.


Subject(s)
Frozen Sections , Lactotrophs/metabolism , Lasers , Microdissection/methods , Pituitary Neoplasms/metabolism , Prolactinoma/metabolism , Proteome/analysis , Proteomics/methods , Cell Separation , Female , Humans , Immunohistochemistry , Lactotrophs/cytology , Lactotrophs/drug effects , Lactotrophs/pathology , Male , Octoxynol/pharmacology , Pituitary Neoplasms/pathology , Prolactinoma/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...