Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Radiat Prot Dosimetry ; 137(3-4): 318-23, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19919960

ABSTRACT

The study of spatial distribution of the indoor radon has assumed in the last years a lot of interest. The geostatistical techniques turn out to be particularly promising. The present work presents the results of a study where around 4000 indoor radon data from Veneto, Friuli Venezia-Giulia and Alto Adige, collected during the sampling campaigns performed in dwellings and in schools, have been analyzed. After the definition of the common data set, the study of the spatial distribution of the phenomenon has been performed by examining the experimental variograms. Declustering techniques have been applied. Predictive maps were defined by using simulation techniques; they allow to determine the probabilities of exceeding defined concentration levels, the 'radon-prone' areas. Systematic results regarding the validation of these maps are reported. This methodological study indicates how it is possible to understand the geographical variability of the phenomenon, trying to find out correlations among indoor radon, geological characteristics (i.e. lithology, morphology, tectonics, soil gas) and building-specific features, which can significantly influence radon concentrations.


Subject(s)
Air Pollution, Indoor/analysis , Air Pollution, Indoor/statistics & numerical data , Air Pollution/analysis , Air Pollution/statistics & numerical data , Data Interpretation, Statistical , Radiation Monitoring/statistics & numerical data , Radon/analysis , Italy , Radiation Dosage
2.
Radiat Prot Dosimetry ; 127(1-4): 287-92, 2007.
Article in English | MEDLINE | ID: mdl-18086689

ABSTRACT

The lung counter facility of the Nuclear Regulatory Authority (ARN) is presented. A calibration was carried out using the Lawrence Livermore National Laboratory (LLNL) phantom. This phantom is provided with a pair of lungs and lymph nodes containing uranium homogeneously distributed and a set of four overlay plates covering a chest wall thickness (CWT) ranging from 1.638 to 3.871 cm. Individual organ calibration factors were acquired for 235U photopeaks energies and for each effective chest thickness. Using these factors, a collection of theoretical fitting curves were found. A counting efficiency formulae and a curve for simultaneously active lymph nodes and lung was obtained and checked through measures. Background measurements of the chamber with and without volunteer persons were performed in order to obtain the detection limits (DL) of the system. As this task involves the knowledge of the volunteers CWTs, these magnitudes were determined through formulae selected from the literature taking into account the detection system characteristics. The deviation in the CWT assigned to an individual, generated by applying different equations, produces variations up to 33% in the estimations of the incorporated activity and DL. An analysis of the changes in efficiencies as consequences of the detectors locations and CWT was also performed. This reveals that the DL of the camera (detectors, shield and blank phantom) is between 2.7 and 6.4 Bq of 235U, which implies 4.9 and 11.5 mg lung burden of natural uranium. An estimation of the minimum detectable intake performed with the DL considering blank persons shows that a system with the characteristics described is only adequate for non-routine individual monitoring.


Subject(s)
Germanium/radiation effects , Government Regulation , Occupational Exposure/analysis , Radiation Monitoring/instrumentation , Radiation Monitoring/standards , Uranium/analysis , Argentina , Equipment Design , Equipment Failure Analysis , Radiation Monitoring/methods , Reproducibility of Results , Sensitivity and Specificity
3.
Biochem Biophys Res Commun ; 285(4): 969-75, 2001 Jul 27.
Article in English | MEDLINE | ID: mdl-11467847

ABSTRACT

Through the use of microarray analysis it was discovered that the nuclear receptor coregulator, receptor interacting protein 140 (RIP140), was induced early during all-trans retinoic acid (RA)-induced differentiation of human embryonal carcinoma cells. A rapid, fourfold induction of RIP140 mRNA was detected within 3 h of RA treatment in human embryonal carcinoma and MCF-7 human breast cancer cells. RIP140 protein levels were induced within 6 h of RA treatment. The RA induction of RIP140 mRNA did not require de novo protein synthesis, consistent with RIP140 being a direct transcriptional target of retinoid receptors. Promoter/enhancer elements directly upstream of the RIP140 coding region supported RA-induced transcription of a luciferase gene. In addition the ability of overexpressed RIP140 to repress ligand activated retinoid receptors was confirmed. The finding that RIP140 is a direct transcriptional target of RA is one of the first examples of acute transcriptional regulation of a nuclear receptor coactivator or corepressor. These data are consistent with a model by which RA induction of RIP140 supplies a negative feedback signal toward ligand-activated retinoid receptors.


Subject(s)
Nuclear Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcriptional Activation , Tretinoin/pharmacology , Adaptor Proteins, Signal Transducing , Breast Neoplasms , Carcinoma, Embryonal , Feedback , Female , Gene Expression Regulation, Neoplastic , Humans , Models, Genetic , Nuclear Receptor Interacting Protein 1 , Teratocarcinoma , Tumor Cells, Cultured
4.
Oncogene ; 20(20): 2559-69, 2001 May 03.
Article in English | MEDLINE | ID: mdl-11420666

ABSTRACT

Although retinoids are known to regulate gene transcription by activating retinoid receptors, the targets of retinoid receptors are largely unknown. This study indicates effective all-trans retinoic acid (RA)-induced differentiation of human embryonal carcinoma cells engages p53. Unexpectedly, RA has been found to activate the transactivation function of p53 in the human embryonal carcinoma cell line, NT2/D1, in a retinoid receptor-dependent manner. A derived RA-resistant line, NT2/D1-R1, is deficient in this activity and is co-resistant to cisplatin. This indicates that RA and cisplatin responses may share a common pathway involving p53 in embryonal carcinomas. RA has no effect on p53 steady-state protein levels in either line. RA enhances endogenous p53 transactivation activity in NT2/D1 but not NT2/D1-R1 cells. In addition, RA induces transactivation activity of a gal4-p53 fusion protein, suggesting that RA activates p53 independent of increasing p53 levels or sequence-specific DNA binding. This activity is absent in retinoic acid receptor gamma (RARgamma)-deficient NT2/D1-R1 cells but can be restored upon co-transfection with specific RARs. Transient transfection of a dominant-negative p53 construct in NT2/D1 cells blocks the RA-mediated transcriptional decline of a differentiation-sensitive reporter plasmid and enhances survival of NT2/D1 cells following cisplatin treatment. Taken together, these findings indicate that RA activates the intrinsic activation function of p53 by a novel mechanism independent of effects on p53 stability or DNA binding and that this activation may be a general mechanism that contributes to RA-mediated G1 arrest.


Subject(s)
Carcinoma, Embryonal/genetics , Gene Expression Regulation, Neoplastic/drug effects , Germinoma/genetics , Testicular Neoplasms/genetics , Transcriptional Activation/drug effects , Tretinoin/pharmacology , Tumor Suppressor Protein p53/physiology , Antineoplastic Agents/pharmacology , Carcinoma, Embryonal/drug therapy , Carcinoma, Embryonal/pathology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Fibroblast Growth Factors/genetics , Genes, p53/drug effects , Genes, p53/genetics , Germinoma/drug therapy , Germinoma/pathology , Humans , Male , Testicular Neoplasms/drug therapy , Testicular Neoplasms/pathology , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
5.
Ann N Y Acad Sci ; 952: 13-22, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11795432

ABSTRACT

The retinoids, natural and synthetic derivatives of vitamin A, are active in cancer therapy and prevention. Their biological effects are mediated through ligand-dependent interactions with retinoid receptors that associate with specific co-regulators. A better understanding of retinoid chemopreventive mechanisms is needed. Our prior work revealed that all-trans-retinoic acid (RA) prevented tobacco-specific carcinogenic transformation of cultured human bronchial epithelial cells. RA signaled G1 arrest that permitted repair of genomic DNA damage caused by these carcinogens. RA triggered G1 arrest at least partly through proteasome-dependent degradation of cyclin D1. Proteasomal inhibitors blocked RA-mediated cyclin D1 degradation. To confirm that a specific proteolysis pathway was induced by RA-treatment, a degradation assay was established using in vitro translated cyclin D1 and cellular extracts from RA-treated or untreated human bronchial epithelial cells. Incubation of RA-treated but not the control cellular extracts with in vitro translated cyclin D1 led to cyclin degradation. This degradation depended on the PEST domain of cyclin D1, implicating ubiquitination in this retinoid degradation. Retinoid receptor selective agonists demonstrated that retinoic acid receptor (RAR)beta and retinoid X receptor (RXR) but not RARalpha- or RARgamma-dependent pathways signaled this cyclin degradation. Findings were extended to the NT2/D1 human embryonal carcinoma differentiation model where a similar pathway was activated by RA-treatment. To determine whether G1 cyclins were involved directly in bronchial preneoplasia, immunohistochemical expression profiles for cyclins D1 and E were examined. Aberrant expression of these cyclins was frequent in bronchial preneoplasia. Taken together, these findings indicate that ubiquitin-dependent proteolysis of G1 cyclins is a retinoid chemoprevention mechanism. Whether the retinoids represent the optimal agents to activate this pathway is the subject of ongoing work. These findings provide a rationale for combining the retinoids in chemoprevention trials with other agents that do not activate this proteolysis pathway. What is now known about the retinoids as cancer prevention agents will be reviewed. Emphasis is placed on retinoid effects on cell cycle progression at G1.


Subject(s)
Anticarcinogenic Agents/pharmacology , Cyclins/metabolism , G1 Phase/drug effects , Neoplasms/prevention & control , Retinoids/pharmacology , Animals , Anticarcinogenic Agents/therapeutic use , Bronchi/cytology , Bronchial Diseases/genetics , Bronchial Diseases/metabolism , Carcinoma, Embryonal/pathology , Cell Differentiation/drug effects , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cysteine Endopeptidases/metabolism , Endopeptidases/metabolism , Epithelial Cells/drug effects , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Metaplasia , Mice , Models, Biological , Multienzyme Complexes/metabolism , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Proteasome Endopeptidase Complex , Protein Processing, Post-Translational , Protein Structure, Tertiary , Receptors, Retinoic Acid/drug effects , Retinoids/therapeutic use , Tretinoin/pharmacology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/pathology , Ubiquitin/metabolism , Vitamin A Deficiency/complications
8.
J Neurol Neurosurg Psychiatry ; 68(1): 83-5, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10601408

ABSTRACT

The alien hand syndrome, as originally defined, was used to describe cases involving anterior corpus callosal lesions producing involuntary movement and a concomitant inability to distinguish the affected hand from an examiner's hand when these were placed in the patient's unaffected hand. In recent years, acceptable usage of the term has broadened considerably, and has been defined as involuntary movement occurring in the context of feelings of estrangement from or personification of the affected limb or its movements. Three varieties of alien hand syndrome have been reported, involving lesions of the corpus callosum alone, the corpus callosum plus dominant medial frontal cortex, and posterior cortical/subcortical areas. A patient with posterior alien hand syndrome of vascular aetiology is reported and the findings are discussed in the light of a conceptualization of posterior alien hand syndrome as a disorder which may be less associated with specific focal neuropathology than are its callosal and callosal-frontal counterparts.


Subject(s)
Attention/physiology , Brain Diseases/physiopathology , Brain/physiopathology , Dominance, Cerebral/physiology , Functional Laterality/physiology , Aged , Brain/pathology , Brain Diseases/pathology , Female , Humans , Magnetic Resonance Imaging , Syndrome
9.
Oncogene ; 18(42): 5747-55, 1999 Oct 14.
Article in English | MEDLINE | ID: mdl-10523855

ABSTRACT

Retinoids signal biological effects through retinoic acid receptors (RAR) and retinoid X receptors (RXR) and their co-regulators. We previously reported that all-trans retinoic acid (RA) triggers terminal differentiation in the human embryonal carcinoma cell line NTERA-2 clone D1 (NT2/D1), through an RARgamma dependent pathway. RARgamma repression in NT2/D1-R1 cells accounts for RA resistance in this line. This report finds RARgamma repression is due to selective repression of RARgamma but not RARbeta transcription in NT2/D1-R1 cells. The repression is neither due to mutations in RARgamma nor its promoter containing the RA response element. Prior work was confirmed and extended by demonstrating that an RARgamma selective agonist preferentially signals differentiation of NT2/D1 cells, while RARalpha/beta, RARbeta, RXR agonists and an RAR pan-antagonist do not even when NT2/D1 cells are treated with these retinoids at 10 microM dosages. None of these examined retinoids induced differentiation of the RA resistant NT2/D1-R1 cells. In contrast, N-(4-hydroxyphenyl)retinamide (4HPR), a reported transcriptional activator of RARgamma was shown to potently induce growth inhibition and apoptosis in both NT2/D1 and NT2/D1-R1 cells. 4HPR-induced apoptosis was unaffected by co-treatment of both cell lines with equimolar RAR antagonist. Semi-quantitative reverse transcription-polymerase chain reaction (RT - PCR) assays of total RNA from 4HPR-treated NT2/D1 and NT2/D1-R1 cells did not reveal RARgamma induction. Since 4HPR signals in RA-resistant NT2/D1-R1 cells having an RARgamma transcriptional block, these results indicate that 4HPR triggers apoptosis but not differentiation through an RARgamma independent pathway. Taken together, these findings implicate a therapeutic role for 4HPR mediated apoptosis in germ cell tumors even when a maturation block is present.


Subject(s)
Apoptosis/drug effects , Carcinoma, Embryonal/metabolism , Fenretinide/pharmacology , Receptors, Retinoic Acid/physiology , Retinoids/metabolism , Anticarcinogenic Agents/pharmacology , Apoptosis/genetics , Carcinoma, Embryonal/genetics , Cell Differentiation/drug effects , DNA Fragmentation/drug effects , DNA Fragmentation/genetics , Humans , Neurons/metabolism , Promoter Regions, Genetic/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/isolation & purification , Receptors, Retinoic Acid/genetics , Teratocarcinoma/genetics , Teratocarcinoma/metabolism , Time Factors , Tretinoin/pharmacology , Tumor Cells, Cultured , Retinoic Acid Receptor gamma
10.
Cell Tissue Res ; 297(3): 371-82, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10460485

ABSTRACT

A novel repressor of the Winged Helix (formerly HNF-3/Forkhead) transcriptional regulatory family, termed Genesis (also called HFH2), was previously found to be exclusively expressed in primitive embryonic cell lines. In this study in situ cRNA hybridization experiments revealed that Genesis was expressed during embryogenesis only in developing neural crest cells. Its expression diminished upon their terminal differentiation into sympathetic and parasympathetic neurons. Based on that finding, Genesis was retrovirally transduced into pluripotent N-Tera-2 clone D1 (NT2/D1) teratocarcinoma cells, which are a well-described in vitro model of neural development. Retinoic acid (RA) treatment will drive these cells to differentiation toward the neuronal lineage and cause an increase in expression of the cyclin-dependent kinase inhibitor p21 protein, which leads to an inhibition in cellular proliferation. Although RA-induced expression of neuronal differentiation markers was not influenced by forced overexpression of Genesis in NT2-D1 cells, proliferation of Genesis-transduced cells continued following RA treatment. RA was unable to induce the expression of the cyclin-dependent kinase inhibitor p21 in the Genesis-transduced cells, but Go/G1 tumor suppressor p53 expression was induced normally. Therefore, Genesis may play a role in the regulation of primitive neural crest development by preventing terminal quiescence through inhibition of p21 protein expression. These data also lend evidence for the hypothesis that proliferation and differentiation pathways are not irrevocably linked, but can function independently.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Neural Crest/embryology , Repressor Proteins/genetics , Animals , Cell Differentiation/genetics , Cell Division/genetics , Embryonic and Fetal Development/genetics , Forkhead Transcription Factors , Helix-Turn-Helix Motifs/genetics , Mice , Organ Specificity , Trans-Activators/genetics
11.
J Biol Chem ; 274(31): 22013-8, 1999 Jul 30.
Article in English | MEDLINE | ID: mdl-10419526

ABSTRACT

Mechanisms by which differentiation programs engage the cell cycle are poorly understood. This study demonstrates that retinoids promote ubiquitination and degradation of cyclin D1 during retinoid-induced differentiation of human embryonal carcinoma cells. In response to all-trans-retinoic acid (RA) treatment, the human embryonal carcinoma cell line NT2/D1 exhibits a progressive decline in cyclin D1 expression beginning when the cells are committed to differentiate, but before onset of terminal neuronal differentiation. The decrease in cyclin D1 protein is tightly associated with the accumulation of hypophosphorylated forms of the retinoblastoma protein and G(1) arrest. In contrast, retinoic acid receptor gamma-deficient NT2/D1-R1 cells do not growth-arrest or accumulate in G(1) and have persistent cyclin D1 overexpression despite RA treatment. Notably, stable transfection of retinoic acid receptor gamma restores RA-mediated growth suppression and differentiation to NT2/D1-R1 cells and restores the decline of cyclin D1. The proteasome inhibitor LLnL blocks this RA-mediated decline in cyclin D1. RA treatment markedly accelerates ubiquitination of wild-type cyclin D1, but not a cyclin D1 (T286A) mutant. Transient expression of cyclin D1 (T286A) in NT2/D1 cells blocks RA-mediated transcriptional decline of a differentiation-sensitive reporter plasmid and represses induction of immunophenotypic neuronal markers. Taken together, these findings strongly implicate RA-mediated degradation of cyclin D1 as a means of coupling induced differentiation and cell cycle control of human embryonal carcinoma cells.


Subject(s)
Cell Cycle/drug effects , Cell Differentiation/physiology , Cyclin D1/metabolism , Protein Processing, Post-Translational/drug effects , Tretinoin/pharmacology , Ubiquitins/metabolism , Carcinoma, Embryonal , Cell Cycle/physiology , Cell Differentiation/drug effects , Cyclin D1/genetics , G1 Phase , Gene Expression Regulation, Neoplastic/drug effects , Humans , Kinetics , Tumor Cells, Cultured
12.
Brain Res ; 829(1-2): 151-9, 1999 May 22.
Article in English | MEDLINE | ID: mdl-10350541

ABSTRACT

Analgesia elicited by morphine in the ventrolateral periaqueductal gray is mediated in part by NMDA and cholinergic receptors in the rostral ventromedial medulla because selective receptor antagonists applied to the latter structure reduced morphine analgesia elicited from the former structure. Previous studies have demonstrated that morphine and beta-endorphin employ different anatomical and neurochemical pathways in exerting their supraspinal analgesic effects. The present study evaluated whether pretreatment with either competitive (AP7, 3-10 microg) or non-competitive (MK-801, 3-10 microg) NMDA antagonists, or muscarinic (scopolamine, 5 microg) or nicotinic (mecamylamine, 1 microg) cholinergic antagonists administered into the rostral ventromedial medulla altered beta-endorphin (15 microg) analgesia elicited from the ventrolateral periaqueductal gray as measured by the tail-flick and jump tests in rats. Whereas AP7 produced minimal (11%) and transient (30 min) reductions in beta-endorphin analgesia on the jump test, MK-801 produced minimal (9%) and transient (30 min) reductions in beta-endorphin analgesia on the tail-flick test. Whereas mecamylamine failed to reduce beta-endorphin analgesia on either measure, scopolamine produced small (23%) and transient (30 min) reductions in beta-endorphin analgesia on the tail-flick test. Each of these antagonists administered into the rostral ventromedial medulla at comparable or lower doses virtually eliminated morphine analgesia elicited from the ventrolateral periaqueductal gray. The opioid mediation of beta-endorphin analgesia in the ventrolateral periaqueductal gray was confirmed by its sensitivity to naltrexone (1-20 microg) pretreatment into the same structure. These data provide further evidence for dissociations between the descending neuroanatomical and neurochemical circuitry mediating the supraspinal analgesic responses induced by morphine and beta-endorphin, and indicate that the latter response is mediated by either non-cholinergic and non-NMDA synapses within the rostral ventromedial medulla, and/or by brainstem sites outside of the rostral ventromedial medulla.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Medulla Oblongata/drug effects , Muscarinic Antagonists/pharmacology , Nicotinic Antagonists/pharmacology , Periaqueductal Gray/drug effects , beta-Endorphin/pharmacology , 2-Amino-5-phosphonovalerate/analogs & derivatives , 2-Amino-5-phosphonovalerate/pharmacology , Analgesia , Animals , Binding, Competitive , Dizocilpine Maleate/pharmacology , Male , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Scopolamine/pharmacology
13.
Cancer Res ; 59(2): 336-41, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-9927043

ABSTRACT

The new synthetic oleanane triterpenoid 2-cyano-3,12-dioxoolean-1,9-dien-28-oic acid (CDDO) is a potent, multifunctional molecule. It induces monocytic differentiation of human myeloid leukemia cells and adipogenic differentiation of mouse 3T3-L1 fibroblasts and enhances the neuronal differentiation of rat PC12 pheochromocytoma cells caused by nerve growth factor. CDDO inhibits proliferation of many human tumor cell lines, including those derived from estrogen receptor-positive and -negative breast carcinomas, myeloid leukemias, and several carcinomas bearing a Smad4 mutation. Furthermore, it suppresses the abilities of various inflammatory cytokines, such as IFN-gamma, interleukin-1, and tumor necrosis factor-alpha, to induce de novo formation of the enzymes inducible nitric oxide synthase (iNos) and inducible cyclooxygenase (COX-2) in mouse peritoneal macrophages, rat brain microglia, and human colon fibroblasts. CDDO will also protect rat brain hippocampal neurons from cell death induced by beta-amyloid. The above activities have been found at concentrations ranging from 10(-6) to 10(-9) M in cell culture, and these results suggest that CDDO needs further study in vivo, for either chemoprevention or chemotherapy of malignancy as well as for neuroprotection.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents/pharmacology , Oleanolic Acid/analogs & derivatives , 3T3 Cells , Animals , Cell Differentiation/drug effects , Cyclooxygenase 2 , Dose-Response Relationship, Drug , Humans , Isoenzymes/drug effects , Membrane Proteins , Mice , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type II , Oleanolic Acid/pharmacology , Prostaglandin-Endoperoxide Synthases/drug effects , Rats
14.
Physiol Behav ; 64(3): 219-25, 1998 Jun 01.
Article in English | MEDLINE | ID: mdl-9748086

ABSTRACT

Serotonergic, NMDA, or opioid antagonists in the rostral ventromedial medulla (RVM) reduce morphine analgesia elicited from the periaqueductal gray (PAG). Continuous (CCWS) and intermittent (ICWS) cold-water swims elicit respective naltrexone-insensitive and naltrexone-sensitive analgesic responses. CCWS analgesia is reduced by systemic NMDA receptor antagonism and by systemic, but not intrathecal serotonergic antagonism. ICWS analgesia is reduced by both systemic and intrathecal serotonergic antagonism, but unaffected by systemic NMDA antagonism. The present study evaluated whether serotonergic (methysergide: 5-10 microg) or competitive [AP7 (2-amino-7-phosphonoheptanoic acid): 0.01-0.1 microg] or non-competitive [MK-801 (dizocilipine maleate): 0.3-3 microg] NMDA antagonists in the RVM altered CCWS and ICWS analgesia and hypothermia as well as basal nociceptive latencies. Methysergide in the RVM significantly potentiated CCWS, but not ICWS analgesia. In contrast, AP7 in the RVM significantly potentiated ICWS analgesia. Antagonist-induced changes in either hypothermia or basal nociception failed to account for any alterations in stress-induced analgesia. These data suggest that serotonergic, but not NMDA, receptors in the RVM may mediate collateral inhibition between mesencephalic morphine analgesia and naltrexone-insensitive CCWS analgesia.


Subject(s)
Hypothermia/physiopathology , Medulla Oblongata/physiology , Pain Threshold/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Serotonin Antagonists/pharmacology , Stress, Physiological/physiopathology , Swimming/physiology , Animals , Hypothermia/etiology , Male , Medulla Oblongata/drug effects , Methysergide/pharmacology , Pain Measurement/drug effects , Rats , Rats, Sprague-Dawley , Stress, Physiological/complications
15.
Oncogene ; 16(26): 3471-80, 1998 Jul 02.
Article in English | MEDLINE | ID: mdl-9692555

ABSTRACT

This study addresses the contributions of specific retinoid receptors during all-trans-retinoic acid (RA)-mediated differentiation and growth suppression of human embryonal carcinoma cells. The pleiotropic effects of RA are mediated by retinoic acid receptors (RARs) and retinoid X receptors (RXRs), members of the nuclear receptor family of transcription factors. After RA-treatment the multipotent human embryonal carcinoma cell line NTERA-2 clone D1 (NT2/D1) displays limited proliferative potential, reduced tumorigenicity, and morphologic and immunophenotypic neuronal maturation. RARgamma over-expression in NT2/D1 cells signals mesenchymal NT2/D1 terminal differentiation while RARalpha and RARbeta do not and RARgamma overcomes retinoid resistance in an NT2/D1 clone (NT2/D1-RI) having deregulated RARgamma expression. Since RARgamma transfectants do not display neuronal maturation, this study sought to identify cooperating retinoid receptors engaged in NT2/D1 differentiation. Through gain of function experiments, this report highlights RXRbeta as playing an important role along with RARgamma in signaling differentiation of NT2/D1 cells. Stable over-expression of RXRbeta, but not RXRalpha or RXRgamma, was found to signal NT2/D1 growth suppression and to induce a non-neuronal morphology and immunophenotype. Notably, co-transfection of RARgamma and RXRbeta resulted in marked growth suppression and for the first time, expression of typical neuronal markers of NT2/D1 differentiation. To clarify the role of RXRbeta and RARgamma in this differentiation program, a modified transient fibroblast growth factor-4 (FGF4) promoter-enhancer reporter assay that reflects effective RA-mediated differentiation of NT2/D1 cells was employed. Transfection of RARgamma or RXRbeta in NT2/D1 cells augments transcriptional repression of the FGF4 reporter and RARgamma and RXRbeta co-transfection markedly repressed reporter activity, indicating the combined role of these receptors in RA-induced NT2/D1 differentiation. Taken together, these findings reveal specific retinoid receptors must cooperate to signal terminal growth suppression and maturation of NT2/D1 cells. Since the transcriptional repression of FGF4 is coupled to the effective maturation of human embryonal carcinoma cells, the described co-transfection strategy should prove useful to identify genes with positive or negative effects on the differentiation program of these tumor cells.


Subject(s)
Carcinoma, Embryonal/metabolism , Receptors, Retinoic Acid/metabolism , Tretinoin/pharmacology , Cell Differentiation , Cell Division , Dimerization , Fibroblast Growth Factor 4 , Fibroblast Growth Factors/biosynthesis , Fibroblast Growth Factors/genetics , Gene Expression Regulation, Neoplastic , Genes, Reporter , Growth Inhibitors , Humans , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Receptors, Retinoic Acid/genetics , Recombinant Proteins/metabolism , Retinoid X Receptors , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Oncogene ; 17(6): 761-7, 1998 Aug 13.
Article in English | MEDLINE | ID: mdl-9715278

ABSTRACT

A subset of male germ cell cancers presenting with advanced stage abundantly express the fibroblast growth factor-4 (FGF4). FGF4 expression is restricted in vitro to undifferentiated embryonal carcinomas (ECs). During induced differentiation, FGF4 expression is repressed in maturation sensitive but not resistant human ECs, suggesting FGF4 plays an important role in malignant growth or differentiation of ECs. To explore these FGF4 signals in male germ cell cancers, the multipotent human EC NTERA-2 clone D1 (NT2/D1) cell line was studied. All-trans-retinoic acid (RA)-treatment of these cells induces a neuronal phenotype and represses tumorigenicity and FGF4 expression. In contrast, RA-treatment of retinoid resistant lines derived from NT2/D1 cells failed to repress FGF4 expression. This implicated FGF4 directly in regulating human EC growth or differentiation. To evaluate further this FGF4 role, FGF4 was constitutively over-expressed in NT2/D1 cells using a CMV-driven expression vector containing the neomycin resistance gene. Three stable transfectants expressing exogenous FGF4 were studied as was a control transfectant only expressing the neomycin resistance gene. RA-treatment repressed endogenous but not exogenous FGF4 expression. RA-treatment of these transfectants induced morphologic and immunophenotypic maturation, changes in RA-regulated genes, and a G1 cell cycle arrest in a manner similar to parental NT2/D1 cells. This indicated FGF4 over-expression did not block RA-mediated differentiation. As expected, RA-treatment repressed tumorigenicity of the control transfectant after subcutaneous injection into athymic mice. Despite RA-treatment, this repressed tumorigenicity was overcome in all the transfectants over-expressing FGF4. The histopathology and neovascularization did not appreciably differ between xenograft tumors derived from FGF4 over-expressing versus control transfectants. FGF4 expression studies were extended to patient-derived germ cell tumors using total cellular RNA Northern analysis and an immunohistochemical assay developed to detect FGF4 protein expression. Germ cell tumors with EC components were significantly more likely to express FGF4 mRNA (P < or = 0.0179) than other examined germ cell tumors without EC components. Immunohistochemical results from 43 germ cell tumors demonstrated increased FGF4 expression especially in non-seminomas having EC components. Thus, FGF4 promotes directly malignant growth of cultured ECs, overcomes the antitumorigenic actions of RA, and is selectively expressed in specific histopathologic subsets of germ cell tumors. Taken together, these findings indicate how differentiation and anti-tumorigenic retinoic acid signals can be dissociated in germ cell cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Embryonal/physiopathology , Fibroblast Growth Factors/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Tretinoin/pharmacology , Cell Cycle , Cell Differentiation/drug effects , Drug Interactions , Fibroblast Growth Factor 4 , Fibroblast Growth Factors/genetics , Humans , Proto-Oncogene Proteins/genetics , Recombinant Proteins/biosynthesis , Signal Transduction , Transfection
17.
Int J Cancer ; 72(6): 1013-20, 1997 Sep 17.
Article in English | MEDLINE | ID: mdl-9378535

ABSTRACT

Melanoma invasion requires migration through the vascular barrier. An early event in this process is the adhesion of metastatic cells to the endothelium. To elucidate the role of TGF-beta in the regulation of this process, human melanoma SK-MEL24 cells were labelled with [5'-(3)H]-thymidine and co-cultured with bovine pulmonary artery endothelial-cell monolayers. Radioactivity was assumed to be proportional to the number of SK-MEL24 cells bound to the endothelium. A low number of melanoma cells adhered to endothelial cells in a time-related manner. Pretreatment for 24 hr with 0.001 to 10 ng/ml TGF-beta1 or TGF-beta2 of both cell types enhanced melanoma-endothelium adhesion in a dose-dependent manner. Both melanoma and endothelial cells expressed RI- and RII-type TGF-beta receptors. The effect of TGF-beta was abolished by co-incubation with the proteoglycan decorin. Conditioned media from melanoma-endothelium co-cultures contained latent TGF-beta and failed to affect cell-cell adhesion. However, activation of TGF-beta by heating the medium or reducing the pH, increased melanoma-endothelium adhesion to an extent similar to that of the TGF-beta administered to the cultures. Zimography demonstrated that both cell types expressed urokinase-type plasminogen activator (uPA). Addition of plasminogen to the co-cultures, which was likely to be activated to plasmin by uPA, resulted in activation of TGF-beta and parallel stimulation of melanoma-endothelium adhesion. In conclusion, TGF-beta may enhance adhesion of melanoma cells to the endothelium, playing a relevant autocrine/paracrine role in the progression of invasive melanoma.


Subject(s)
Activin Receptors, Type I , Cell Adhesion/drug effects , Endothelium, Vascular/physiology , Melanoma/physiopathology , Skin Neoplasms/physiopathology , Transforming Growth Factor beta/pharmacology , Animals , Cattle , Cells, Cultured , Coculture Techniques , Decorin , Endothelium, Vascular/cytology , Extracellular Matrix Proteins , Fibrinolysin/metabolism , Humans , Kinetics , Melanoma/pathology , Plasminogen/metabolism , Protein Serine-Threonine Kinases/biosynthesis , Proteoglycans/pharmacology , Pulmonary Artery , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/biosynthesis , Recombinant Proteins/pharmacology , Skin Neoplasms/pathology , Tumor Cells, Cultured , Urokinase-Type Plasminogen Activator/biosynthesis
18.
Biochim Biophys Acta ; 1353(2): 191-8, 1997 Aug 07.
Article in English | MEDLINE | ID: mdl-9294013

ABSTRACT

RT-PCR analysis of the reduced folate carrier (RFC) from L1210 and murine erythroleukemia cells led to the identification of three clones which appeared to result from the use of alternative splice sites. The nucleotide sequence of each splice form predicts a protein that contains at least the first 7 transmembrane domains of the parental RFC protein followed by a novel hydrophilic carboxyl terminus of 33, 72, or 105 amino acid residues. Sequence analysis of cDNA clones isolated from murine liver and the results of 5'-RACE from L1210 cells indicated that RFC also utilizes alternate 5'-terminal exons. To understand how the alternatively spliced RFC transcripts and multiple 5'-termini were generated, the genomic organization of RFC was determined. The gene is comprised of at least 8 exons, the first two of which encode the alternative 5' termini. Based on sequence identity with cDNAs encoding RFC from hamster and rat, however, it appears that additional 5' exons may be present. Two of the RFC splice variants result from the use of a cryptic splice donor site within exon 4 and the third results from the use of a cryptic splice acceptor site within exon 5. In addition, the splice variant form that encodes the largest protein also utilizes an alternative exon located between exons 5 and 6. The apparent use of alternative transcriptional start sites and the identification of several RFC splice forms raises the possibility that unique RFC molecules may be generated that exhibit tissue- or cell line-specific distribution.


Subject(s)
Alternative Splicing , Carrier Proteins/genetics , Membrane Proteins , Membrane Transport Proteins , Animals , Base Sequence , Carrier Proteins/chemistry , Cloning, Molecular , DNA, Complementary/chemistry , DNA, Complementary/isolation & purification , Exons , Genomic Library , Liver/metabolism , Mice , Molecular Sequence Data , Polymerase Chain Reaction , Reduced Folate Carrier Protein , Tumor Cells, Cultured
19.
J Mol Endocrinol ; 18(2): 167-76, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9134503

ABSTRACT

We have cloned a cDNA encoding the full-length coding region of the human homologue of the germ cell nuclear factor (GCNF)/retinoid receptor-related testis-associated receptor (RTR), from a human testis cDNA library. The amino acid sequence of human GCNF/RTR is highly homologous to that of the mouse GCNF/RTR. The largest difference between the two homologues is a 15 amino acid deletion in the human GCNF/RTR at amino acid 47. The GCNF/RTR gene was localized on human chromosome 9. Northern blot analysis using poly(A)+ RNA from different human tissues showed that GCNF/RTR mRNA is most abundantly expressed in the testis. GCNF/RTR was also highly expressed in embryonic stem cells and embryonal carcinoma cells but repressed in its differentiated derivatives. Induction of differentiation of mouse embryonal carcinoma F9 cells and human embryonal carcinoma NTERA-2 clone D1 (NT2/D1) cells by all-trans retinoic acid was accompanied by a down-regulation of GCNF/RTR. Our observations suggest that GCNF/RTR plays a role in the control of gene expression in early embryogenesis and during spermatogenesis.


Subject(s)
Cloning, Molecular , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Amino Acid Sequence , Animals , Base Sequence , Cell Differentiation/genetics , Chromosome Mapping , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , DNA-Binding Proteins/biosynthesis , Embryonal Carcinoma Stem Cells , Humans , Male , Mice , Molecular Sequence Data , Neoplastic Stem Cells/chemistry , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/metabolism , Nuclear Receptor Subfamily 6, Group A, Member 1 , RNA, Messenger/chemistry , RNA, Messenger/genetics , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Retinoic Acid/biosynthesis , Stem Cells/chemistry , Stem Cells/cytology , Stem Cells/metabolism
20.
Biochem Pharmacol ; 53(2): 223-31, 1997 Jan 24.
Article in English | MEDLINE | ID: mdl-9037255

ABSTRACT

F2-MTX'A is an L1210 leukemia cell line with a functional detect in the reduced folate carrier and high level expression of folate receptor beta. The pH dependence of methotrexate (MTX) influx by folate receptor beta in F2-MTX'A cells was characterized and compared with that of the reduced folate carrier in parental L1210 cells. MTX influx by folate receptor beta had a pH optimum of 6.5, whereas influx mediated by the reduced folate carrier showed a pH optimum of 7.5. Increased folate receptor beta-mediated MTX influx at pH 6.5 relative to pH 7.5 was accompanied by a 5-fold increase in binding affinity of the receptor for MTX without a change in the number of binding sites. At pH 6.2, approximately 24% of MTX influx in F2-MTX'A cells proceeded by another mechanism. This transport route became active at pH < 7.5, operated optimally at pH 6.0 to 6.5, and, unlike folate receptor beta-mediated MTX influx, was insensitive to the presence of low levels of folic acid (100 nM). MTX influx by the low pH system showed saturability, with a Ki of 5.3 microM and a Vmax of 1.53 nmol/g dry wt/min, was energy dependent, was inhibited by sulfobromophthalein with a Ki of 148 microM, and had similar relative affinities for folic acid, leucovorin, and 5 methyltetrahydrofolate. Influx of 5-methyltetrahydrofolate was also mediated by this route. The data provide further confirmatory evidence for an MTX influx route in F2-MTX'A cells, optimal at low pH and distinct from the reduced folate carrier or the folate receptor.


Subject(s)
Carrier Proteins/physiology , Folic Acid Antagonists/pharmacokinetics , Leukemia L1210/metabolism , Membrane Proteins , Membrane Transport Proteins , Methotrexate/pharmacokinetics , Receptors, Cell Surface , Animals , Biological Transport , Cell Division/drug effects , Folate Receptors, GPI-Anchored , Hydrogen-Ion Concentration , Methotrexate/pharmacology , Mice , Reduced Folate Carrier Protein , Sulfobromophthalein/pharmacology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...