Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Clin Endocrinol (Oxf) ; 73(4): 452-6, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20626412

ABSTRACT

OBJECTIVE: To assess whether clinician-determined treatment intervention thresholds are in line with the assessment of fracture risk provided by FRAX® and treatment recommendations provided by UK guidelines produced by the National Osteoporosis Guidelines Group (NOGG). DESIGN, PATIENTS AND MEASUREMENTS: This was a retrospective cohort analysis of 288 patients consecutively referred for dual-energy X-ray absorptiometry (DXA) scanning from primary care immediately prior to the introduction of the FRAX® algorithm. In addition to DXA assessment, patients completed a clinical risk factor questionnaire which included risk factors used in the FRAX® algorithm. Initial risk assessment and treatment decisions were performed after DXA. FRAX® was used, retrospectively, with femoral neck T-score, to estimate fracture risk which was applied to NOGG to generate guidance on treatment intervention. Clinician- and NOGG-determined outcomes were audited for concordance. RESULTS: There was concordance between clinician and NOGG treatment decisions in 215 (74.6%) subjects. Discordance was observed in 73 (25.3%) subjects. In the discordant group, seven subjects were given lifestyle advice when NOGG recommended treatment, 42 given treatment when NOGG recommended lifestyle advice only, and 24 were referred to a metabolic bone clinic for further evaluation. The reasons for treatment differences in subjects recommended treatment by clinician but not NOGG were largely (90.2%) attributed to the use of lumbar spine bone mineral density (BMD). CONCLUSIONS: There is high concordance between clinician-determined and FRAX®-NOGG intervention. The absence of spine BMD from FRAX® is the primary source of discrepancy. This study provides some assurance of the validity of the treatment thresholds generated from FRAX®-NOGG in 'real-world' usage.


Subject(s)
Osteoporosis/therapy , Absorptiometry, Photon , Aged , Algorithms , Female , Humans , Male , Middle Aged , Practice Guidelines as Topic , Retrospective Studies , United Kingdom
2.
Clin Med (Lond) ; 10(1): 45-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20408307

ABSTRACT

Most patients with primary hyperparathyroidism (PHPT) are asymptomatic at presentation. This presents the dilemma whether to treat surgically or manage by conservative follow-up. This article covers the risks of managing mild PHPT conservatively. Some of these risks are well established, for example worsening of bone disease and increased risk of nephrolithiasis. Others, such as effects on cardiovascular function or the risk of malignancy are more controversial. These factors are critical to decisions relating to surgical or conservative management of mild PHPT.


Subject(s)
Hyperparathyroidism, Primary/complications , Hyperparathyroidism, Primary/therapy , Bone Diseases/etiology , Bone Diseases/pathology , Bone Diseases/prevention & control , Cardiovascular Diseases/etiology , Cardiovascular Diseases/pathology , Cardiovascular Diseases/prevention & control , Humans , Hyperparathyroidism, Primary/diagnosis , Parathyroidectomy , Risk Factors
3.
J Mol Endocrinol ; 33(3): 663-77, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15591026

ABSTRACT

Human pituitary tumor-transforming gene (PTTG), known also as securin, is a multifunctional protein implicated in the control of mitosis and the pathogenesis of thyroid, colon, oesophageal and other tumour types. Critical to PTTG function is a C-terminal double PXXP motif, forming a putative SH3-interacting domain and housing the gene's sole reported phosphorylation site. The exact role of phosphorylation and PXXP structure in the modulation of PTTG action in vitro remains poorly understood. We therefore examined the mitotic, transformation, proliferation and transactivation function of the C-terminal PXXP motifs of human PTTG. Live-cell imaging studies using an EGFP-PTTG construct indicated that PTTG's regulation of mitosis is retained regardless of phosphorylation status. Colony-formation assays demonstrated that phosphorylation of PTTG may act as a potent inhibitor of cell transformation. In proliferation assays, NIH-3T3 cells stable transfected and overexpressing mutations preventing PTTG phosphorylation (Phos-) showed significantly increased [3H]thymidine incorporation compared with WT, whereas mutants mimicking constitutive phosphorylation of PTTG (Phos+) exhibited reduced cell proliferation. We demonstrated that PTTG transactivation of FGF-2 in primary thyroid and PTTG-null cell lines was not affected by PTTG phosphorylation but was prevented by a mutant disrupting the PXXP motifs (SH3-). Taken together, our data suggest that PXXP structure and phosphorylation are likely to exert independent and critical influences upon PTTG's diverse actions in vitro.


Subject(s)
Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Amino Acid Motifs , Animals , Cell Proliferation/drug effects , Cell Transformation, Neoplastic , Cells, Cultured , Fibroblast Growth Factor 2/pharmacology , Humans , Mice , Mutation/genetics , Neoplasm Proteins/genetics , Phosphorylation/drug effects , Securin
4.
FASEB J ; 17(12): 1631-9, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12958169

ABSTRACT

Human securin, known also as PTTG, has established oncogenic and cell cycle regulatory functions. PTTG/securin transforms cells in vitro, inhibits sister chromatid separation, and regulates secretion of fibroblast growth factor-2. FGF-2 is a key regulator of CNS development and PTTG/securin expression has been reported in murine fetal brain. We examined the expression and function of securin and FGF-2 in the developing human fetal brain and in a fetal neuronal cell line (NT 2). Securin expression was significantly reduced in first and second trimester fetal cerebral cortex compared with adult cerebral cortex, where immunocytochemistry revealed intense securin staining in neuronal cell bodies. FGF-2 protein was concordantly lower in fetal cortex, whereas pretranslational expression of PTTG binding factor (PBF) was not significantly altered in fetal brain compared with adult. PCNA expression demonstrated that high securin levels in adult cortex were associated with absent cell proliferation. In NT-2 cells, securin stimulated FGF-2 expression, which could be abrogated by a carboxyl-terminal mutation. Low transient expression of securin resulted in a significant proliferative effect, whereas high levels of securin expression inhibited cell turnover. We propose a potential role for human PTTG/securin in modulating cell proliferation and FGF-2 expression during human neurogenesis.


Subject(s)
Brain/embryology , Membrane Proteins , Neoplasm Proteins/physiology , Brain/cytology , Brain/metabolism , Brain Chemistry , Cell Division , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryonic and Fetal Development , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Neoplasm Proteins/analysis , Neoplasm Proteins/genetics , Neurons/metabolism , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Fibroblast Growth Factor, Type 1 , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Securin , Up-Regulation
5.
J Steroid Biochem Mol Biol ; 85(2-5): 415-21, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12943730

ABSTRACT

The enzymes 11beta-hydroxysteroid dehydrogenase type 1 and 2 (11beta-HSD1 and 2) have well-defined roles in the tissue-specific metabolism of glucocorticoids which underpin key endocrine mechanisms such as adipocyte differentiation (11beta-HSD1) and mineralocorticoid action (11beta-HSD2). However, in recent studies we have shown that the effects of 11beta-HSD1 and 2 are not restricted to distinct tissue-specific hormonal functions. Studies of normal fetal and adult tissues, as well as their tumor equivalents, have shown a further dichotomy in 11beta-HSD expression and activity. Specifically, most normal glucocorticoid receptor (GR)-rich tissues such as adipose tissue, bone, and pituitary cells express 11beta-HSD1, whereas their fetal equivalents and tumors express 11beta-HSD2. We have therefore postulated that the ability of 11beta-HSD1 to generate cortisol acts as an autocrine anti-proliferative, pro-differentiation stimulus in normal adult tissues. In contrast, the cortisol-inactivating properties of 11beta-HSD2 lead to pro-proliferative effects, particularly in tumors. This proposal is supported by experiments in vitro which have demonstrated divergent effects of 11beta-HSD1 and 2 on cell proliferation. Current studies are aimed at (1) characterizing the underlying mechanisms for a "switch" in 11beta-HSD isozyme expression in tumors; (2) defining the molecular targets for glucocorticoids as regulators of cell proliferation; (3) evaluating the potential for targeting glucocorticoid metabolism as therapy for some cancers. These and other issues are discussed in the present review.


Subject(s)
Cell Division/physiology , Cell Transformation, Neoplastic , Hydroxysteroid Dehydrogenases/metabolism , Neoplasms/enzymology , 11-beta-Hydroxysteroid Dehydrogenases , Animals , Humans , Isoenzymes/metabolism , Models, Biological
6.
J Clin Endocrinol Metab ; 88(5): 2341-7, 2003 May.
Article in English | MEDLINE | ID: mdl-12727994

ABSTRACT

Differentiated thyroid cancers are the most common endocrine cancers, but there are no reliable molecular markers of prognosis. Pituitary tumor transforming gene (PTTG) plays several potential roles in tumor initiation and progression, including regulating mitosis and stimulating expression of fibroblast growth factor (FGF)-2. Increased expression of PTTG has been demonstrated in follicular thyroid lesions, and expression of this oncogene has been identified as a potential prognostic marker in pituitary adenomas and colon carcinomas. We assessed the expression of PTTG and FGF-2 and its receptor FGF-R-1 in 27 differentiated thyroid cancers, and we compared this with expression in 11 normal thyroids, 25 multinodular goiters, and 13 Graves' disease specimens. We also examined the relationship between gene expression and clinical markers of tumor behavior. PTTG and FGF-2 were overexpressed in thyroid carcinomas (9.5-fold increase, P = 0.003, and 5.0-fold increase, P < 0.001, respectively) compared with normal thyroid. Increased FGF-2 mRNA expression was independently associated with the findings of lymph node invasion (R(2) = 0.71; P < 0.001) and distant metastasis (R(2) = 0.55; P = 0.009) at tumor presentation, after taking into account known prognostic factors such as age and gender of the patient and size and type of the tumor. High PTTG expression was independently associated with tumor recurrence (R(2) = 0.64; P = 0.003). We conclude that PTTG and FGF-2 expression are potential prognostic markers (and perhaps therapeutic targets) for differentiated thyroid cancer.


Subject(s)
Fibroblast Growth Factor 2/genetics , Gene Expression , Neoplasm Proteins/genetics , Thyroid Neoplasms/genetics , Adult , Biomarkers, Tumor/analysis , Female , Goiter, Nodular/metabolism , Graves Disease/metabolism , Humans , Male , Neoplasm Recurrence, Local , Prognosis , Proliferating Cell Nuclear Antigen/genetics , RNA, Messenger/analysis , Receptors, Fibroblast Growth Factor/genetics , Securin , Thyroid Gland/chemistry
7.
Oncogene ; 22(11): 1663-7, 2003 Mar 20.
Article in English | MEDLINE | ID: mdl-12642869

ABSTRACT

The physiological effects of glucocorticoids (GCs) are, at least in part, mediated by inhibition of cell proliferation. Two isozymes of 11 beta-hydroxysteroid dehydrogenase (11 beta-HSD) interconvert cortisol (F) and inactive cortisone (E), and are thus able to modulate GC action at an autocrine level. Previously, we have demonstrated absent expression of 11 beta-HSD2 in normal pituitaries; however, in a small number of pituitary tumors analysed, 11 beta-HSD2 was readily demonstrable. Here we have used real-time RT-PCR to quantify expression of mRNA for 11 beta-HSD1 and 2 in 105 human pituitary tumors and have performed enzyme expression and activity studies in primary pituitary cultures. Overall, pituitary tumors expressed lower levels of 11 beta-HSDl mRNA compared with normals (0.2-fold, P<0.05). In contrast, expression of 11 beta-HSD2 mRNA was 9.8-fold greater in tumors than in normals (P<0.001). Enzyme assays showed significant 11 beta-HSD2 activity (71.9+/-22.3 pmol/h/mg protein (mean+/-s.d.)) but no detectable 11 beta-HSDl activity. Proliferation assays showed that addition of glycyrrhetinic acid (an 11 beta-HSD2 inhibitor) resulted in a 30.3+/-7.7% inhibition of cell proliferation. In summary, we describe a switch in expression from 11 beta-HSDl to 11 beta-HSD2 in neoplastic pituitary tissue. We propose that abnormal expression of 11 beta-HSD2 acts as a proproliferative prereceptor determinant of pituitary cell growth, and may provide a novel target for future tumor therapy.


Subject(s)
Adenoma/enzymology , Cell Division , Hydroxysteroid Dehydrogenases/genetics , Pituitary Neoplasms/enzymology , 11-beta-Hydroxysteroid Dehydrogenases , Adenoma/pathology , Base Sequence , DNA Primers , Humans , Pituitary Neoplasms/pathology , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
8.
Clin Endocrinol (Oxf) ; 58(2): 141-50, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12580928

ABSTRACT

OBJECTIVE: Pituitary tumour transforming gene (PTTG) encodes a multifunctional protein that is implicated in initiating and perpetuating pituitary adenoma growth. PTTG appears to have key regulatory functions in determining control of many fundamental cellular events including mitosis, cell transformation, DNA repair and gene regulation. Several of these events are mediated through interactions with PTTG binding factor (PBF) and fibroblast growth factor-2 (FGF-2). Given this background, we have determined the expression of PTTG, PBF, FGF-2 and its receptor FGF-R-1 in a large cohort of pituitary adenomas and have sought associations between levels of gene expression and clinical markers of tumour behaviour. PATIENTS AND METHODS: We used real-time reverse transcriptase-polymerase chain reaction (RT-PCR) and Western blot analyses to measure PTTG, PBF, FGF-2 and FGF-R-1 expression in ex vivo pituitary tumours (N = 121). Clinical data, including accurate radiological assessment of tumour characteristics, were used to determine any associations between gene expression and tumour behaviour. RESULTS: PTTG was increased significantly (fivefold, P = 0.005) in adenomas compared with normal pituitaries. We also demonstrated that PBF was similarly raised in adenomas (sixfold, P = 0.0001), and was significantly correlated with PTTG expression. FGF-2 and its receptor FGF-R-1 were also raised in adenomas compared with normal pituitary tissue. Moreover, significantly enhanced expression of FGF-R-1 was observed in invasive adenomas compared with other pituitary tumours. CONCLUSIONS: Our data support a fundamental role for PTTG-mediated upregulation of FGF-2 signalling in pituitary tumorigenesis and growth, and suggest that receptor-mediated mechanisms of growth factor action may be critically important. Further prospective studies are required to determine whether measurement of FGF-R-1 mRNA will be of clinical use as a prognostic marker in patients with pituitary adenomas.


Subject(s)
Adenoma/chemistry , Biomarkers, Tumor/analysis , Fibroblast Growth Factor 2/analysis , Membrane Proteins , Neoplasm Proteins/genetics , Pituitary Neoplasms/chemistry , Adenoma/pathology , Adult , Blotting, Western/methods , Chi-Square Distribution , Cohort Studies , DNA-Binding Proteins/analysis , DNA-Binding Proteins/genetics , Female , Fibroblast Growth Factor 2/genetics , Gene Expression , Humans , Intracellular Signaling Peptides and Proteins , Magnetic Resonance Imaging , Male , Middle Aged , Neoplasm Proteins/analysis , Pituitary Neoplasms/pathology , RNA, Messenger/analysis , Receptor Protein-Tyrosine Kinases/analysis , Receptor Protein-Tyrosine Kinases/genetics , Receptor, Fibroblast Growth Factor, Type 1 , Receptors, Fibroblast Growth Factor/analysis , Receptors, Fibroblast Growth Factor/genetics , Reverse Transcriptase Polymerase Chain Reaction , Securin , Statistics, Nonparametric
9.
J Clin Endocrinol Metab ; 87(9): 4238-44, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12213878

ABSTRACT

Pituitary tumorigenesis is a poorly understood process involving dysregulation of the cell cycle, proliferation, and angiogenesis. The novel securin pituitary tumor transforming gene (PTTG) disrupts cell division and stimulates fibroblast growth factor (FGF)-2-mediated angiogenesis. We investigated expression of the angiogenic vascular endothelial growth factor (VEGF) and its receptor KDR/Flk-1 in 103 human pituitary tumors, and we assessed functional relationships between these genes in vitro. Nonfunctioning tumors (n = 81) demonstrated markedly raised VEGF mRNA (3.2-fold, P < 0.05) and protein concentrations, compared with normal pituitaries (n = 10). KDR was also highly induced in nonfunctioning tumors (14-fold, P < 0.001, n = 78) as well as in the whole cohort of pituitary tumors, compared with normal pituitary samples (14-fold, P < 0.0001, n = 100). In vitro, PTTG induced VEGF, but not KDR, expression in fetal neuronal NT2 cells (2.7-fold, P < 0.001, n = 8), MCF-7 breast carcinoma cells (1.9-fold, P = 0.03, n = 10), and choriocarcinoma JEG-3 cells (P = 0.0002, n = 8). A mutated PTTG construct that cannot be phosphorylated showed identical VEGF up-regulation (2.9-fold, P < 0.001, n = 8) in NT2 cells, compared with wild-type PTTG, but a further mutated construct with abrogation of the key protein:protein interaction domain of PTTG resulted in a significant reduction in VEGF stimulation, compared with wild-type (0.37-fold reduction, P < 0.001, n = 8). FGF-2 findings mirrored those of VEGF, although antibody depletion of secreted FGF-2 in the cell medium failed to influence VEGF up-regulation by PTTG. Overall, our findings implicate altered VEGF and KDR signaling in pituitary tumorigenesis, and we propose that PTTG stimulation of FGF-2 and VEGF expression in the presence of up-regulated growth factor receptors may account for angiogenic growth and progression of human pituitary tumors.


Subject(s)
Endothelial Growth Factors/genetics , Gene Expression Regulation, Neoplastic , Lymphokines/genetics , Neoplasm Proteins/genetics , Pituitary Neoplasms/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Growth Factor/genetics , Transcription, Genetic , Adenoma/blood supply , Adenoma/genetics , Adenoma/surgery , Amino Acid Substitution , Base Sequence , DNA Primers , Humans , Mutagenesis, Site-Directed , Neovascularization, Pathologic/genetics , Pituitary Gland/metabolism , Pituitary Neoplasms/blood supply , Pituitary Neoplasms/surgery , Polymerase Chain Reaction , RNA, Messenger/genetics , Receptors, Vascular Endothelial Growth Factor , Recombinant Proteins/metabolism , Regression Analysis , Securin , Trans-Activators/genetics , Transfection , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
10.
Clin Endocrinol (Oxf) ; 56(6): 735-43, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12072042

ABSTRACT

OBJECTIVE: Thyroid hormones (THs) perform essential roles in pituitary function. They regulate anterior pituitary hormone secretion and are also key determinants of pituitary cell proliferation and differentiation. The critical role of deiodinase enzymes, which serve as prereceptor regulators of TH action, remains largely unexplored. Three deiodinase enzymes metabolize active and inactive THs and thereby determine tissue concentrations of the biologically active ligand, tri-iodothyronine (T3). We hypothesized that aberrant expression of deiodinase enzymes and/or altered enzyme activity in pituitary tumours may change tissue concentrations of THs and influence their growth and secretory characteristics. STUDY DESIGN AND PATIENTS: We studied 105 pituitary tumours and 10 normal pituitaries for expression of deiodinase enzyme mRNAs encoding types 1 (D1), 2 (D2) and 3 (D3) using real-time RT-PCR. Enzyme activity data from 20 pituitary samples were also obtained. RESULTS: Pituitary tumours expressed significantly increased D3 mRNA (6.5-fold, P < 0.0005) compared with normal pituitaries. D2 mRNA was also increased 2.6-fold (P = 0.005) in pituitary tumours compared with normals. The rare TSH-secreting pituitary tumour subtype expressed a 13.1-fold excess of D3 mRNA and reduced D2 mRNA (0.1-fold of normal pituitaries). D2 mRNA expression in ACTH-secreting tumours was similarly reduced to 0.1-fold that in normal pituitaries. CONCLUSIONS: Pituitary adenomas express abnormal levels of deiodinase enzymes compared to normal pituitaries. These abnormalities may have functional consequences on pituitary tumour growth. In the case of TSH-secreting pituitary adenomas, the observed pattern of deiodinase mRNA expression may explain the 'resistance' of this tumour type to TH feedback.


Subject(s)
Adenoma/enzymology , Iodide Peroxidase/genetics , Isoenzymes/genetics , Pituitary Neoplasms/enzymology , Adenoma/metabolism , Adrenocorticotropic Hormone/metabolism , Adult , Humans , Iodide Peroxidase/metabolism , Isoenzymes/metabolism , Middle Aged , Pituitary Gland/enzymology , Pituitary Neoplasms/metabolism , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Thyrotropin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...