Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Neoplasia ; 43: 100926, 2023 09.
Article in English | MEDLINE | ID: mdl-37597490

ABSTRACT

The Sonic Hedgehog (Hh) signal transduction pathway plays a critical role in many developmental processes and, when deregulated, may contribute to several cancers, including basal cell carcinoma, medulloblastoma, colorectal, prostate, and pancreatic cancer. In recent years, several Hh inhibitors have been developed, mainly acting on the Smo receptor. However, drug resistance due to Smo mutations or non-canonical Hh pathway activation highlights the need to identify further mechanisms of Hh pathway modulation. Among these, deacetylation of the Hh transcription factor Gli1 by the histone deacetylase HDAC1 increases Hh activity. On the other end, the KCASH family of oncosuppressors binds HDAC1, leading to its ubiquitination and subsequent proteasomal degradation, leaving Gli1 acetylated and not active. It was recently demonstrated that the potassium channel containing protein KCTD15 is able to interact with KCASH2 protein and stabilize it, enhancing its effect on HDAC1 and Hh pathway. KCTD15 and KCTD1 proteins share a high homology and are clustered in a specific KCTD subfamily. We characterize here KCTD1 role on the Hh pathway. Therefore, we demonstrated KCTD1 interaction with KCASH1 and KCASH2 proteins, and its role in their stabilization by reducing their ubiquitination and proteasome-mediated degradation. Consequently, KCTD1 expression reduces HDAC1 protein levels and Hh/Gli1 activity, inhibiting Hh dependent cell proliferation in Hh tumour cells. Furthermore, analysis of expression data on publicly available databases indicates that KCTD1 expression is reduced in Hh dependent MB samples, compared to normal cerebella, suggesting that KCTD1 may represent a new putative target for therapeutic approaches against Hh-dependent tumour.


Subject(s)
Cerebellar Neoplasms , Hedgehog Proteins , Male , Humans , Hedgehog Proteins/genetics , Zinc Finger Protein GLI1/genetics , Cell Proliferation , Databases, Factual , Co-Repressor Proteins
2.
Oncogene ; 36(32): 4641-4652, 2017 08 10.
Article in English | MEDLINE | ID: mdl-28368412

ABSTRACT

Aberrant Hedgehog/GLI signaling has been implicated in a diverse spectrum of human cancers, but its role in lung adenocarcinoma (LAC) is still under debate. We show that the downstream effector of the Hedgehog pathway, GLI1, is expressed in 76% of LACs, but in roughly half of these tumors, the canonical pathway activator, Smoothened, is expressed at low levels, possibly owing to epigenetic silencing. In LAC cells including the cancer stem cell compartment, we show that GLI1 is activated noncanonically by MAPK/ERK signaling. Different mechanisms can trigger the MAPK/ERK/GLI1 cascade including KRAS mutation and stimulation of NRP2 by VEGF produced by the cancer cells themselves in an autocrine loop or by stromal cells as paracrine cross talk. Suppression of GLI1, by silencing or drug-mediated, inhibits LAC cells proliferation, attenuates their stemness and increases their susceptibility to apoptosis in vitro and in vivo. These findings provide insight into the growth of LACs and point to GLI1 as a downstream effector for oncogenic pathways. Thus, strategies involving direct inhibition of GLI1 may be useful in the treatment of LACs.


Subject(s)
Adenocarcinoma/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Zinc Finger Protein GLI1/metabolism , Adenocarcinoma/pathology , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Female , Humans , Lung Neoplasms/pathology , Mice , Mice, SCID , Mitogen-Activated Protein Kinase Kinases/metabolism , Neoplastic Stem Cells/pathology , Neuropilin-2/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Pyridines/pharmacology , Pyrimidines/pharmacology , RNA Interference/physiology , RNA, Small Interfering/metabolism , Xenograft Model Antitumor Assays , Zinc Finger Protein GLI1/antagonists & inhibitors , Zinc Finger Protein GLI1/genetics
3.
Cell Death Differ ; 20(12): 1688-97, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24013724

ABSTRACT

The Hedgehog (Hh) signaling regulates tissue development, and its aberrant activation is a leading cause of malignancies, including medulloblastoma (Mb). Hh-dependent tumorigenesis often occurs in synergy with other mechanisms, such as loss of p53, the master regulator of the DNA damage response. To date, little is known about mechanisms connecting DNA-damaging events to morphogen-dependent processes. Here, we show that genotoxic stress triggers a cascade of signals, culminating with inhibition of the activity of Gli1, the final transcriptional effector of Hh signaling. This inhibition is dependent on the p53-mediated elevation of the acetyltransferase p300/CBP-associated factor (PCAF). Notably, we identify PCAF as a novel E3 ubiquitin ligase of Gli1. Indeed PCAF, but not a mutant with a deletion of its ubiquitination domain, represses Hh signaling in response to DNA damage by promoting Gli1 ubiquitination and its proteasome-dependent degradation. Restoring Gli1 levels rescues the growth arrest and apoptosis effect triggered by genotoxic drugs. Consistently, DNA-damaging agents fail to inhibit Gli1 activity in the absence of either p53 or PCAF. Finally, Mb samples from p53-null mice display low levels of PCAF and upregulation of Gli1 in vivo, suggesting PCAF as potential therapeutic target in Hh-dependent tumors. Together, our data define a mechanism of inactivation of a morphogenic signaling in response to genotoxic stress and unveil a p53/PCAF/Gli1 circuitry centered on PCAF that limits Gli1-enhanced mitogenic and prosurvival response.


Subject(s)
DNA Damage , Kruppel-Like Transcription Factors/metabolism , Signal Transduction , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , p300-CBP Transcription Factors/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , HEK293 Cells , Hedgehog Proteins/metabolism , Humans , Kruppel-Like Transcription Factors/chemistry , Mice , Mitogens/pharmacology , Models, Biological , Proteolysis/drug effects , Signal Transduction/drug effects , Transcription Factors/chemistry , Ubiquitination/drug effects , Zinc Finger Protein GLI1
4.
Oncogene ; 31(33): 3807-17, 2012 Aug 16.
Article in English | MEDLINE | ID: mdl-22120716

ABSTRACT

Post-translational modifications of Notch3 and their functional role with respect to Notch3 overexpression in T-cell leukemia are still poorly understood. We identify here a specific novel property of Notch3 that is acetylated and deacetylated at lysines 1692 and 1731 by p300 and HDAC1, respectively, a balance impaired by HDAC inhibitors (HDACi) that favor hyperacetylation. By using HDACi and a non-acetylatable Notch3 mutant carrying K/R(1692-1731) mutations in the intracellular domain, we show that Notch3 acetylation primes ubiquitination and proteasomal-mediated degradation of the protein. As a consequence, Notch3 protein expression and its transcriptional activity are decreased both in vitro and in vivo in Notch3 transgenic (tg) mice, thus impairing downstream signaling upon target genes. Consistently, Notch3-induced T-cell proliferation is inhibited by HDACi, whereas it is enhanced by the non-acetylatable Notch3-K/R(1692-1731) mutant. Finally, HDACi-induced Notch3 hyperacetylation prevents in vivo growth of T-cell leukemia/lymphoma in Notch3 tg mice. Together, our findings suggest a novel level of Notch signaling control in which Notch3 acetylation/deacetylation process represents a key regulatory switch, thus representing a suitable druggable target for Notch3-sustained T-cell acute lymphoblastic leukemia therapy.


Subject(s)
Leukemia, T-Cell/etiology , Receptors, Notch/physiology , Acetylation , Animals , HEK293 Cells , Histone Deacetylase Inhibitors/therapeutic use , Humans , Leukemia, T-Cell/drug therapy , Lymphocyte Activation , Mice , Proteasome Endopeptidase Complex/physiology , Receptor, Notch3 , T-Lymphocytes/immunology , Ubiquitination
5.
Oncogene ; 30(1): 65-76, 2011 Jan 06.
Article in English | MEDLINE | ID: mdl-20818436

ABSTRACT

Hedgehog pathway regulates tissue patterning and cell proliferation. Gli1 transcription factor is the major effector of Hedgehog signaling and its deregulation is often associated to medulloblastoma formation. Proteolytic processes represent a critical mechanism by which this pathway is turned off. Here, we characterize the regulation of an ubiquitin-mediated mechanism of Gli1 degradation, promoted by the coordinated action of the E3 ligase Itch and the adaptor protein Numb. We show that Numb activates the catalytic activity of Itch, releasing it from an inhibitory intramolecular interaction between its homologous to E6-AP C-terminus and WW domains. The consequent activation of Itch, together with the recruitment of Gli1 through direct binding with Numb, allows Gli1 to enter into the complex, resulting in Gli1 ubiquitination and degradation. This process is mediated by a novel Itch-dependent degron, composed of a combination of two PPXYs and a phospho-serine/proline motifs, localized in Gli1 C-terminal region, indicating the role of two different WW docking sites in Gli1 ubiquitination. Remarkably, Gli1 protein mutated in these modules is no longer regulated by Itch and Numb, and determines enhanced Gli1-dependent medulloblastoma growth, migration and invasion abilities, as well as in vitro transforming activity. Our data reveal a novel mechanism of regulation of Gli1 stability and function, which influences Hedgehog/Gli1 oncogenic potential.


Subject(s)
Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Hedgehog Proteins/metabolism , Humans , Mice , NIH 3T3 Cells , Repressor Proteins/metabolism , Signal Transduction , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Zinc Finger Protein GLI1
6.
Horm Metab Res ; 35(8): 498-501, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12953168

ABSTRACT

Hyper- and hypothyroidism have significant effects on the female reproductive system. However, little in the way of data is available on the relationship between ovarian paracrine control and thyroid function. This study was aimed at characterising the serum levels of inhibin B in relation to altered thyroid function. Serum inhibin B and FSH levels were measured in 91 women (51 regularly cycling and 40 postmenopausal). The mean serum concentration of inhibin B in euthyroid cycling women (0.025 +/- 0.018 microg/l) was similar to that observed in hyper- and hypothyroid patients (0.022 +/- 0.015 and 0.018 +/- 0.014 microg/l, respectively, p=ns). Inhibin B levels were obviously reduced (-72%) in euthyroid postmenopausal women. In contrast, in hyper- and hypothyroid postmenopausal women, inhibin B levels remained substantially at the premenopausal level. So far, serum inhibin B appeared to be significantly increased in both hyperthyroid patients (0.025 +/- 0.014 microg/l; p<0.0001) and in hypothyroid patients (0.016 +/- 0.006 microg/l; p=0.0006). Altered thyroid function did not affect FSH levels at fertile age. However, a significant decrease of FSH levels was observed in hyper- and hypothyroid (-52% and -43%, respectively) postmenopausal women. Nevertheless, these FSH levels remained in the postmenopausal range. These results indicate that an altered thyroid function affects serum inhibin B levels in postmenopausal women.


Subject(s)
Hyperthyroidism/physiopathology , Hypothyroidism/physiopathology , Inhibins/blood , Postmenopause/blood , Thyroid Gland/physiopathology , Adult , Case-Control Studies , Female , Follicle Stimulating Hormone/blood , Humans , Menstrual Cycle/blood , Middle Aged , Osmolar Concentration , Thyroid Function Tests
7.
J Endocrinol Invest ; 25(11): 967-70, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12553556

ABSTRACT

The aim of this study was to analyze the serum levels of activin A in hyperthyroid patients with Graves' disease. Serum activin A and FSH levels were measured in a total of 93 females (64 regularly cycling and 29 post-menopausal). Of these, 20 were hyperthyroid patients with Graves disease, 33 were euthyroid goitrous patients (20 had autoimmune thyroiditis AT and 13 only had goiter) representing the internal control group and 40 were healthy subjects representing the external control group. Serum levels of activin A were higher in goitrous patients with AT than in control subjects (p=0.0388). Activin A levels were almost doubled in the cycling and in post-menopausal hyperthyroid women (0.91+/-0.21 vs 0.43+/-0.07 microg/l; p<0.0001 and 0.92+/-0.22 vs 0.48+/-0.24 microg/l; p=0.0001, respectively). In 10 cycling hyperthyroid patients, studied even after methimazole treatment, that increase was substantially reversed, once euthyroidism was attained (p=0.002). These findings indicate that thyroid function and autoimmune processes significantly affect serum levels of activin A in patients with Graves' disease.


Subject(s)
Activins/blood , Graves Disease , Graves Disease/blood , Inhibin-beta Subunits/blood , Adult , Aged , Autoantibodies/blood , Female , Follicle Stimulating Hormone/blood , Graves Disease/drug therapy , Graves Disease/immunology , Humans , Methimazole/therapeutic use , Middle Aged , Postmenopause , Thyrotropin/blood , Thyroxine/blood
8.
Proc Natl Acad Sci U S A ; 98(19): 10936-41, 2001 Sep 11.
Article in English | MEDLINE | ID: mdl-11535812

ABSTRACT

Ser-133 phosphorylation of the cAMP-responsive element-binding protein (CREB) is sufficient to induce cellular gene expression in response to cAMP, but additional promoter-bound factors are required for target gene activation by CREB in response to mitogen/stress signals. To compare the relative effects of different signals on recruitment of the coactivator CREB-binding protein (CBP) to CREB in living cells, we developed a fluorescence resonance energy transfer (FRET) assay. cAMP promoted the interaction of CREB with CBP in a phosphorylation-dependent manner by FRET analysis, but mitogen/stress signals were far less effective in stimulating complex formation even though they induced comparable levels of Ser-133 phosphorylation. cAMP and non-cAMP stimuli were comparably active in promoting this interaction in the cytosol; the formation of CREB x CBP complexes in response to non-cAMP signals was specifically inhibited in the nucleus. Non-cAMP signals had no effect on intrinsic CREB- or CBP-binding activities by Far Western blot assay, thereby supporting the presence of a distinct CREB x CBP antagonist. Our studies indicate that the relative effects of cAMP and mitogen/stress signals on CREB x CBP complex formation impart selectivity to gene activation through CREB phosphorylated at Ser-133.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP/metabolism , Mitogens/metabolism , Nuclear Proteins/metabolism , Signal Transduction , Tetradecanoylphorbol Acetate/metabolism , Trans-Activators/metabolism , Animals , CREB-Binding Protein , Cell Line , Gene Expression Regulation , Humans , Mitogens/pharmacology , PC12 Cells , Phosphorylation , Rats , Serine/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transcriptional Activation
9.
Eur J Endocrinol ; 143(2): 267-71, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10913947

ABSTRACT

OBJECTIVE: The selenoenzyme type 2 iodothyronine 5' deiodinase (DII) catalyzes the conversion of thyroxine into its active form tri-iodothyronine (T3), modulating thyroid hormone homeostasis in a local, tissue-specific manner. The amphibian, rodent and human cDNAs encoding this enzyme have been recently cloned and expressed. At present, little information regarding the genomic structure of mammalian DII is available. DESIGN AND METHODS: The complete structure, including intron-exon junctions, of the human DII (hDII) gene was obtained by long PCR and rapid amplification of cDNA ends (RACE). Chromosomal assignment of the hDII gene was performed by fluorescence in situ hybridization using a highly specific probe. RESULTS AND CONCLUSIONS: Our data demonstrated that hDII is a single copy gene located on chromosome 14, position 14q24.3. The gene spans over 15 kb, and the 7 kb transcript is encoded by three exons of 149 bp, 273 bp and 6.6 kb separated respectively by two 274 bp and 7.4 kb introns. A restriction map of the hDII gene is also reported. These data will help in further studies of the role of DII in the maintenance of peripheral thyroid hormone homeostasis.


Subject(s)
Chromosome Mapping , DNA, Complementary/chemistry , Iodide Peroxidase/genetics , Alternative Splicing , Base Sequence , Chromosomes, Human, Pair 14 , Exons , Humans , In Situ Hybridization, Fluorescence , Introns , Molecular Sequence Data , Polymerase Chain Reaction , RNA, Messenger/genetics , Restriction Mapping , Sequence Homology
10.
Eur J Endocrinol ; 143(1): 119-23, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10870040

ABSTRACT

OBJECTIVE: We have studied the effect of tryptophan on cellular [(125)I]tri-iodothyronine (T3) uptake by mouse thymocytes. MATERIALS AND METHODS: Mouse thymocytes (20 x 10(6 )cells/ml) were suspended in Krebs-Ringer solution buffered by Tris-HCl and incubation (23 degrees C at pH7.45+/-0.6), in the presence or absence of 1mM tryptophan, was started by adding 25 pM [(125)I]T3. At the end of incubation, samples were cooled in ice, centrifuged over a 30% sucrose cushion and the cell-associated radioactivity was measured in the pellet. RESULTS: Tryptophan reduced both the total and the saturable fraction of [(125)I]T3 uptake by 44% (P=0.0009) and 60% (P=0.0006) respectively, following 1 min of incubation. This effect was specific and dose-dependent, being maximal at 5mM concentration (-82%). In contrast, the pre-exposure of cells to tryptophan for up to 2h had no effect on the subsequent uptake of [(125)I]T3, in the absence of tryptophan. The effect of D-tryptophan on saturable T3 uptake was not different from that obtained using the L-stereoisomer. Tryptophan reduced the V(max) of the initial rate of saturable [(125)I]T3 uptake by two-thirds without affecting the apparent K(m) (2.2 nM) of the process, thus indicating the non-competitive nature of the inhibition. In sodium-free medium the saturable [(125)I]T3 uptake was reduced by 43%. The inhibitory effect of tryptophan on [(125)I]T3 uptake was exerted in both the presence and the absence of sodium. In fact, the inhibitory effect of tryptophan on T3 transport was greater and significantly different (P=0.0046) from that obtained by sodium depletion alone. CONCLUSIONS: Tryptophan interferes with both the sodium-dependent and -independent components of [(125)I]T3 uptake by a dose-dependent, non-competitive mechanism which operates in cis-modality at the plasma membrane level of mouse thymocytes.


Subject(s)
Thymus Gland/drug effects , Thymus Gland/metabolism , Triiodothyronine/metabolism , Tryptophan/pharmacology , Animals , Biological Transport/drug effects , Iodine Radioisotopes , Kinetics , Mice , Mice, Inbred BALB C , Sodium/pharmacology
11.
Endocrinology ; 141(5): 1804-13, 2000 May.
Article in English | MEDLINE | ID: mdl-10803591

ABSTRACT

We analyzed the structure and function of the 5' flanking region of the human type 2 deiodinase (hD2) gene. Two major transcription start sites were identified at -470/-474 from the ATG. The 5' flanking region of hD2 gene efficiently directed transcription in transient transfection studies, using luciferase as reporter gene, in HEK 293 cells. Basal transcriptional activity was significantly reduced by deleting the region containing a canonical cAMP-responsive element (CRE) located -766/-759 from ATG. Forskolin treatment significantly increased luciferase activity in cells transfected with CRE-containing constructs. This effect was abolished in constructs that did not contain CRE or contained the mutagenized CRE. Northern blot analysis in JEG-3 cells revealed that the hD2 messenger RNA was markedly increased after stimulation with cAMP agonist. The electrophoretic mobility shift assay with hD2-CRE probe and HEK 293 nuclear extract showed the occurrence of a DNA-protein complex, which was competed by specific unlabeled oligonucleotides and supershifted by the anti-CREB and anti-CRE modulator-1 antibodies. A-CREB, a dominant negative inhibitor of CREB, completely inhibited forskolin induction of the hD2 promoter. CREB protein, once cotransfected with hD2 promoter construct and pKA in F9 teratocarcinoma cells, which are unresponsive to cAMP, was able to stimulate the hD2 gene transcription. These results indicate the existence of a functional promoter within the 5' flanking region of hD2 gene which is characterized by the presence of a CRE. The specific involvement of CREB in the cAMP-mediated hD2 gene promoter induction also has been demonstrated.


Subject(s)
Cyclic AMP/metabolism , Iodide Peroxidase/genetics , Promoter Regions, Genetic , Base Sequence , Blotting, Northern , Cell Line , Colforsin/pharmacology , Cyclic AMP Response Element-Binding Protein/antagonists & inhibitors , Cyclic AMP Response Element-Binding Protein/metabolism , Electrophoresis, Polyacrylamide Gel , Humans , Molecular Sequence Data , Numerical Analysis, Computer-Assisted , Structure-Activity Relationship , Teratoma/metabolism , Tumor Cells, Cultured , Iodothyronine Deiodinase Type II
12.
Mol Cell Endocrinol ; 141(1-2): 49-52, 1998 Jun 25.
Article in English | MEDLINE | ID: mdl-9723885

ABSTRACT

Type II 5'-Deiodinase (5'DII) is a key element in the maintenance of peripheral thyroid hormone homeostasis through the regulation of local T4 to T3 conversion in pituitary, brain, brown adipose tissue and placenta. The cDNA containing the coding region of the human 5'DII (HDII) has been recently cloned from infant brain. In the present paper we report the genomic structure, chromosomal localization and restriction map of the coding region of HDII. The presence of a single intron located at codon 75 was demonstrated using a PCR-based strategy; the exon-intron junctions were then cloned and partially sequenced. Chromosomal localization was performed by radiation hybrid mapping. This study demonstrated that the entire coding region of the HDII gene is contained in two exons spliced at codon 75 by a 7.4 Kb intron and that the HDII chromosomal location is 14q24.3. These data will allow further studies of the role of HDII in the pathophysiology of thyroid homeostasis.


Subject(s)
Chromosome Mapping , Iodide Peroxidase/genetics , Open Reading Frames/genetics , Base Sequence , Exons , Humans , Hybrid Cells , Introns , Molecular Sequence Data , Polymerase Chain Reaction , Restriction Mapping , Iodothyronine Deiodinase Type II
13.
Thyroid ; 8(2): 155-9, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9510124

ABSTRACT

The relation between thyroid homeostasis and the biochemical parameters of subclinical protein malnutrition has been analyzed in schoolchildren in a rural area in the south of Italy, known to be moderately iodine-deficient. The sera of 32 children (15 males and 17 females aged 6 to 11 years) have been analyzed. These children were divided into two groups, according to thyroid function: (1) 16 euthyroid children (mean thyrotropin [TSH] 2.38 +/- .35 mU/L; 6 with goiter) and (2) 16 subclinical hypothyroid children (mean TSH 7.32 +/- 1.68 mU/L; 6 with goiter). Retinol circulating complex (RCC) components were determined in serum by high-performance liquid chromatography (HPLC) and radial immunodiffusion and the essential and nonessential amino acid levels by ion exchange chromatography. Reduced retinol binding protein (RBP) and transthyretin (TTR) levels were recorded in the sera of 11 of 32 (34%) and in 5 of 32 (16%) patients, respectively. The linear regression analysis revealed that RBP and TSH levels were inversely correlated (r = -0.514; p < 0.0026). The RBP levels were subnormal in 2 of 16 euthyroid and in 9 of 16 hypothyroid patients (Fisher test p < 0.023), and the mean RBP levels were significantly reduced in the hypothyroid patients when compared with those of the euthyroid group (p < 0.0026). The retinol/RBP ratio was also significantly different between euthyroid and hypothyroid children (0.75 vs. 0.95; p < 0.0002). The mean essential amino acid levels, with the exception of methionine, were all in the normal range. The selected amino acid ratios confirmed that the patients were exposed to mild protein malnutrition. These results provide evidence that even mild protein-energy malnutrition may have detrimental effects on thyroid homeostasis in iodine-deficient areas.


Subject(s)
Child Nutrition Disorders/physiopathology , Homeostasis/physiology , Iodine/deficiency , Protein-Energy Malnutrition/complications , Protein-Energy Malnutrition/physiopathology , Thyroid Gland/physiopathology , Amino Acids/blood , Child , Child Nutrition Disorders/blood , Female , Goiter/blood , Humans , Hypothyroidism/blood , Male , Prealbumin/analysis , Protein-Energy Malnutrition/blood , Reference Values , Retinol-Binding Proteins/analysis , Thyrotropin/blood , Vitamin A/blood
14.
Eur J Endocrinol ; 136(6): 595-8, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9225722

ABSTRACT

OBJECTIVE: The aim of the study was to analyse the relationship between the ocular parameters, namely intraocular pressure (IOP), and the early forms of subclinical hypothyroidism. DESIGN: Fifty-three subjects (9 male and 44 female) aged from 18 to 45 years (mean 32 +/- 7 years) were selected for this study. Twenty-nine met the criteria of subclinical hypothyroidism and 24 euthyroid subjects, age- and sex-matched, were used as controls. METHODS: All individuals underwent a complete ocular examination, including visual field examination and serial measurement of IOP by means of a Goldmann tonometer. A tonographic examination was also performed. RESULTS: The hypothyroid patients showed a substantially higher pressure in both eyes compared with control subjects (right eye = 17.52 +/- 4.74 vs 13.42 +/- 1.95 mmHg, P < 0.0001; left eye = 17.55 +/- 3.99 vs 13.71 +/- 1.55 mmHg, P < 0.0001). Indeed, the tonometric pressure exceeded 18 mmHg in 11 out of the 29 (38%) patients in the right eye and in 8 out of 29 (27%) patients in the left eye. The outflow index was normal in all subjects except in two hypothyroid patients. After two months of L-thyroxine (L-T4) replacement therapy, only one patient continued to show tonometric values above 18 mmHg and the hypothyroid patients showed a significant reduction in mean IOP in both eyes compared with pre-treatment values (right eye = 14.96 +/- 1.32 mmHg, P < 0.0097; left eye = 15.03 +/- 1.38 mmHg, P < 0.0018). Treatment did not lead to any change in the outflow indices; however, the C value (outflow coefficient at the sclerocorneal corner) returned to normal in the two patients with increased pre-treatment tonographic values. CONCLUSIONS: These findings indicate that the intraocular pressure is increased even in subclinical hypothyroid patients and that, at this early stage, the impairment is fully reversible with L-T4 therapy.


Subject(s)
Hypothyroidism/physiopathology , Intraocular Pressure , Adolescent , Adult , Female , Humans , Hypothyroidism/drug therapy , Intraocular Pressure/drug effects , Male , Middle Aged , Reference Values , Thyroxine/therapeutic use , Tonometry, Ocular
15.
Eur J Endocrinol ; 135(6): 696-9, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9025715

ABSTRACT

The aim of this study has been to investigate the plasma endothelin-1 (ET-1) levels in adult patients with proven Addison's disease (AD). Plasma ET-1 levels were measured in 29 subjects (17 males and 12 females, aged between 20 and 54 years): 15 of them were patients with AD and 14 were sex- and age-matched normal subjects, used as a control group. All patients with AD have been studied under basal conditions and nine of them also after 2 weeks on oral corticosteroid therapy (individual cortisol dosage ranging from 25 to 37.5 mg/day and 0.1 mg/day 9 alpha-fluorohydrocortisone). Extracted plasma ET-1 was determined by a specific radioimmunoassay using rabbit endothelin antisera. Mean ET-1 values in the patients with AD were three times higher than in normal subjects (21.09 +/- 4.38 pg/ml vs 6.72 +/- 1.74 pg/ml; p < 0.0001). Plasma ET-1 levels assayed in the patients with AD after 2 weeks of corticosteroid therapy were significantly decreased (14.47 +/- 3.7 pg/ml vs 22.8 +/- 5.2 pg/ml; -37%; p < 0.001) compared to values in untreated patients. However, the plasma ET-1 values obtained following corticosteroid therapy were still significantly higher (p < 0.001) than those detected in the control subjects. These results clearly indicate that patients with untreated AD have increased circulating ET-1 levels that may be reduced by short-term corticosteroid therapy.


Subject(s)
Addison Disease/blood , Endothelin-1/blood , Addison Disease/drug therapy , Addison Disease/physiopathology , Adult , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Endothelin-1/drug effects , Endothelin-1/immunology , Female , Fludrocortisone/pharmacology , Fludrocortisone/therapeutic use , Hemodynamics/drug effects , Humans , Hydrocortisone/pharmacology , Hydrocortisone/therapeutic use , Immune Sera/immunology , Male , Middle Aged , Mineralocorticoids/pharmacology , Mineralocorticoids/therapeutic use , Rabbits , Radioimmunoassay , Reference Values
SELECTION OF CITATIONS
SEARCH DETAIL
...