Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Rev Esp Enferm Dig ; 100(8): 481-9, 2008 Aug.
Article in Spanish | MEDLINE | ID: mdl-18942901

ABSTRACT

OBJECTIVE: To assess the effect of pentoxiphylline (a potent inhibitor of tumor necrosis factor alpha) on survival, on systemic and portal hemodynamics, and on cardiac function in patients with alcoholic cirrhosis. DESIGN: A randomized double-blind placebo-controlled trial. SETTING: A single center using parallel groups of patients to compare pentoxiphylline with placebo. PATIENTS: We recruited 24 patients with alcoholic cirrhosis (8 Child-Pugh B and 16 Child-Pugh C). INTERVENTIONS: Patients were randomly assigned to receive pentoxiphylline (400 mg tid; n = 12) or placebo (n = 12) over a 4-week period. OUTCOME MEASURES: The primary outcome was to extend short-term and long-term survival. Secondary outcomes included hemodynamic benefits (improvement in cardiac function and/or systemic vascular resistance index, or decrease in portal pressure). RESULTS: Portal pressure and cardiac function remained unchanged and there were no significant differences in short-term or long-term survival between treatment and placebo groups. The group on pentoxiphylline increased systemic vascular resistance and decreased cardiac indices (from 1,721 +/- 567 to 2,082 +/- 622 dyn.sec(-1) cm(-5) m(-2) and from 4.17 +/- 1.4 to 3.4 +/- 0.9 l.m(-2), p = 0.05). CONCLUSIONS: Although pentoxiphylline seems to provide some short-term hemodynamic benefits in patients with advanced alcoholic cirrhosis, this drug has no effect on survival or portal pressure in these patients.


Subject(s)
Liver Cirrhosis, Alcoholic/drug therapy , Liver Cirrhosis, Alcoholic/physiopathology , Pentoxifylline/therapeutic use , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Double-Blind Method , Female , Heart/drug effects , Heart/physiopathology , Hemodynamics/drug effects , Humans , Liver Cirrhosis, Alcoholic/mortality , Male , Middle Aged , Portal System/drug effects , Portal System/physiopathology , Severity of Illness Index , Survival Rate
2.
Rev. esp. enferm. dig ; 100(8): 481-489, ago. 2008. ilus, tab
Article in Es | IBECS | ID: ibc-71015

ABSTRACT

Objetivo: valorar el efecto de la pentoxifilina (un potente inhibidordel factor de necrosis tumoral alfa) en la supervivencia, en lahemodinámica sistémica y portal y en la función cardiaca en la cirrosisalcohólica avanzada.Diseño: estudio aleatorizado, doble-ciego, controlado con placebo.Contexto: estudio unicéntrico utilizando grupos de pacientesen paralelo para comparar pentoxifilina y placebo.Pacientes: se incluyeron 24 pacientes con cirrosis alcohólica(8 en estadio B de Child-Pugh y 16 en estadio C de Child-Pugh).Intervención: los pacientes fueron aleatorizados a recibirpentoxifilina (400 mg, 3 veces al día, n = 12) o placebo (n = 12)durante 4 semanas.Determinaciones: el objetivo principal fue la supervivencia acorto/largo plazo. Los objetivos secundarios fueron observar beneficioshemodinámicos (mejoría en la función cardiaca y/o en elíndice de resistencias vasculares sistémicas o disminución de lapresión portal).Resultados: la presión portal y la función cardiaca no se modificarony no hubo diferencias en la supervivencia a corto y largoplazo entre los grupos tratados y placebo. Los índices de resistenciavascular sistémica y cardiaco cambiaron en el grupo de pentoxifilina(de 1.721 ± 567 a 2.082 ± 622 Din.seg1 cm-5 m-2 y de4,17 ± 1,4 a 3,4 ± 0,9 lm-2, p = 0,05).Conclusiones: aunque la pentoxifilina parece producir algúnbeneficio hemodinámico a corto plazo en pacientes con cirrosis alcohólicaavanzada, no tiene efecto sobre la tasa de supervivencia, lafunción cardiaca ni sobre la presión portal en estos pacientes


Objective: to assess the effect of pentoxiphylline (a potent inhibitorof tumor necrosis factor alpha) on survival, on systemicand portal hemodynamics, and on cardiac function in patientswith alcoholic cirrhosis.Design: a randomized double-blind placebo-controlled trial.Setting: a single center using parallel groups of patients tocompare pentoxiphylline with placebo.Patients: we recruited 24 patients with alcoholic cirrhosis (8Child-Pugh B and 16 Child-Pugh C).Interventions: patients were randomly assigned to receivepentoxiphylline (400 mg tid; n = 12) or placebo (n = 12) over a 4-week period.Outcome measures: the primary outcome was to extendshort-term and long-term survival. Secondary outcomes includedhemodynamic benefits (improvement in cardiac function and/orsystemic vascular resistance index, or decrease in portal pressure).Results: portal pressure and cardiac function remained unchangedand there were no significant differences in short-term orlong-term survival between treatment and placebo groups. Thegroup on pentoxiphylline increased systemic vascular resistanceand decreased cardiac indices (from 1,721 ± 567 to 2,082 ± 622dyn.sec-1 cm-5 m-2 and from 4.17 ± 1.4 to 3.4 ± 0.9 l.m-2, p =0.05).Conclusions: although pentoxiphylline seems to providesome short-term aemodynamic benefits in patients with advancedalcoholic cirrhosis, this drug has no effect on survival or portalpressure in these patients


Subject(s)
Humans , Male , Middle Aged , Liver Cirrhosis, Alcoholic/drug therapy , Liver Cirrhosis, Alcoholic/physiopathology , Pentoxifylline/therapeutic use , Tumor Necrosis Factor-alpha/agonists , Double-Blind Method , Heart , Heart/physiopathology , Liver Cirrhosis, Alcoholic/mortality , Portal System , Portal System/physiopathology , Severity of Illness Index , Survival Rate
3.
Neuroscience ; 153(3): 687-99, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18400407

ABSTRACT

There is recent evidence supporting the notion that the cannabinoid signaling system plays a modulatory role in the regulation of cell proliferation and migration, survival of neural progenitors, neuritic elongation and guidance, and synaptogenesis. This assumption is based on the fact that cannabinoid 1-type receptors (CB(1) receptors) and their ligands emerge early in brain development and are abundantly expressed in certain brain regions that play key roles in these processes. We have recently presented in vivo evidence showing that this modulatory action might be exerted through regulating the synthesis of the cell adhesion molecule L1 that is also a key element for those processes. To further explore this issue, we conducted here immunohistochemical studies aimed at determining the cellular substrates of CB(1) receptor-L1 interactions in the rat brain during late fetal development. In this period, we previously found that the activation of CB(1) receptors increased L1 synthesis in several forebrain white matter regions but not in gray matter areas. Using double labeling studies, we observed here colocalization of both proteins in fiber tracts including the corpus callosum, the adjacent subcortical white matter, the internal capsule and the anterior commissure. Experiments conducted with cultures of fetal rat cortical nerve cells revealed that L1 is present mainly in neurons but not in glial cells. This fact, together with the results obtained in the double labeling studies, would indicate that L1 and CB(1) receptors should possibly be present in axons elongating through these white matter tracts, or, alternatively, in migrating neurons. Further experiments confirmed the presence of CB(1) receptors in elongating axons, since these receptors colocalized with growth-associated protein 43 (GAP-43), a marker of growth cones, but not with synaptophysin, a marker of active synaptic terminals, in the same forebrain white matter regions. Lastly, using cultured fetal rat cortical neurons, we also observed that the activation of cannabinoid receptors increased the levels of the full-length L1 and altered those of some active proteolytic fragments of this protein whose generation has been associated with specific steps in the process of neuritic elongation in cultured neurons. In summary, we have demonstrated that the effects caused by cannabinoid agonists on L1 are facilitated by the colocalization of this cell adhesion molecule with CB(1) receptors in several forebrain white matter regions during fetal brain development. We have provided strong evidence that this phenomenon occurs in axons elongating through these white matter tracts, and we have explored in vitro how cannabinoid receptors influence L1 levels. Considering the role played by L1 in different events related to neural development, our observations support the occurrence of a physiological mechanism by which the cannabinoid system might regulate the process of axonal growth and guidance through regulating the synthesis and function of L1.


Subject(s)
GAP-43 Protein/metabolism , Growth Cones/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Prosencephalon/embryology , Prosencephalon/metabolism , Receptor, Cannabinoid, CB1/metabolism , Animals , Blotting, Western , Female , Fetus , Fluorescent Antibody Technique , Immunohistochemistry , Rats , Rats, Wistar , Synaptophysin/metabolism
4.
Br J Pharmacol ; 153(2): 277-85, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17934510

ABSTRACT

The presence of functional cannabinoid CB2 receptors in the CNS has provoked considerable controversy over the past few years. Formerly considered as an exclusively peripheral receptor, it is now accepted that it is also present in limited amounts and distinct locations in the brain of several animal species, including humans. Furthermore, the inducible nature of these receptors under neuroinflammatory conditions, in contrast to CB1, makes them attractive targets for the development of novel therapeutic approaches. In fact, the undesired psychoactive effects caused by CB1 activation have largely limited the clinical use of cannabinoid-related compounds that act on these receptors. In this review some recent findings on the antiinflammatory properties of CB2 receptors are presented, as well as new perspectives that have been obtained based on studies of human postmortem brain samples. In addition, various working hypotheses are also proposed and discussed.


Subject(s)
Encephalitis/physiopathology , Receptor, Cannabinoid, CB2/physiology , AIDS Dementia Complex/pathology , AIDS Dementia Complex/physiopathology , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Encephalitis/pathology , Humans , Inflammation/pathology , Inflammation/physiopathology , Receptor, Cannabinoid, CB2/agonists
5.
Neuroscience ; 151(1): 104-10, 2008 Jan 02.
Article in English | MEDLINE | ID: mdl-18068305

ABSTRACT

Recent data suggest that the endocannabinoid system (ECS) may be involved in the glial response in different types of brain injury. Both acute and chronic insults seem to trigger a shift in the pattern of expression of some elements of this system from neuronal to glial. Specifically, data obtained in human brain tissue sections from Alzheimer's disease patients showed that the expression of cannabinoid receptors of the CB(2) type is induced in activated microglial cells while fatty acid amide hydrolase (FAAH) expression is increased in reactive astrocytes. The present study was designed to determine the time-course of the shift from neuronal to glial induction in the expression of these proteins in Down's syndrome, sometimes referred to as a human model of Alzheimer-like beta-amyloid (Abeta) deposition. Here we present immunohistochemical evidence that both CB(2) receptors and FAAH enzyme are induced in Abeta plaque-associated microglia and astroglia, respectively, in Down's syndrome. These results suggest that the induction of these elements of the ECS contributes to, or is a result of, amyloid deposition and subsequent plaque formation. In addition, they confirm a striking differential pattern of distribution of FAAH and CB(2) receptors.


Subject(s)
Amidohydrolases/metabolism , Amyloid beta-Peptides/physiology , Down Syndrome/metabolism , Neuroglia/metabolism , Receptor, Cannabinoid, CB2/metabolism , Adult , Brain/pathology , Brain Chemistry/physiology , Child , Down Syndrome/pathology , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Infant, Newborn , Male , Middle Aged , Plaque, Amyloid/pathology
6.
Life Sci ; 75(16): 1907-15, 2004 Sep 03.
Article in English | MEDLINE | ID: mdl-15306158

ABSTRACT

The role of the endocannabinoid system in several diseases is currently under intense study. Among these, Alzheimer's disease may be a new promising area of research. We have recently reported the existence of profound changes in the location and density of several elements of this system in Alzheimer's disease tissue samples, indicating that a non-neuronal endocannabinoid system is up-regulated in activated glia. Additional data from other groups suggest that glial cells may be important elements in the regulation of endocannabinoid system activity, both in health as in disease. Some of these aspects are briefly discussed in the present review.


Subject(s)
Alzheimer Disease/physiopathology , Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Neuroglia/metabolism , Alzheimer Disease/metabolism , Amidohydrolases/metabolism , Humans , Models, Biological , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism
7.
Mol Cell Biol ; 20(23): 8793-802, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11073980

ABSTRACT

The vitamin D receptor (VDR) normally functions as a ligand-dependent transcriptional activator. Here we show that, in the presence of Ets-1, VDR stimulates the prolactin promoter in a ligand-independent manner, behaving as a constitutive activator. Mutations in the AF2 domain abolish vitamin D-dependent transactivation but do not affect constitutive activation by Ets-1. Therefore, in contrast with the actions of vitamin D, activation by Ets-1 is independent of the AF2 domain. Ets-1 also conferred a ligand-independent activation to the estrogen receptor and to peroxisome proliferator-activated receptor alpha. In addition, Ets-1 cooperated with the unliganded receptors to stimulate the activity of reporter constructs containing consensus response elements fused to the thymidine kinase promoter. There is a direct interaction of the receptors with Ets-1 which requires the DNA binding domains of both proteins. Interaction with Ets-1 induces a conformational change in VDR which can be detected by an increased resistance to proteolytic digestion. Furthermore, a retinoid X receptor-VDR heterodimer in which both receptors lack the core C-terminal AF2 domain can recruit coactivators in the presence, but not in the absence, of Ets-1. This suggests that Ets-1 induces a conformational change in the receptor which creates an active interaction surface with coactivators even in the AF2-defective mutants. These results demonstrate the existence of a novel mechanism, alternative to ligand binding, which can convert an unliganded receptor from an inactive state into a competent transcriptional activator.


Subject(s)
Proto-Oncogene Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Transcriptional Activation , CREB-Binding Protein , Estrogens/metabolism , Genes, Reporter , Histone Acetyltransferases , Ligands , Nuclear Proteins/metabolism , Nuclear Receptor Coactivator 1 , Prolactin/genetics , Promoter Regions, Genetic , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Proto-Oncogene Protein c-ets-1 , Proto-Oncogene Proteins c-ets , Receptors, Calcitriol/metabolism , Receptors, Estrogen/metabolism , Response Elements , Trans-Activators/metabolism , Vitamin D/metabolism
8.
Brain Res ; 868(2): 338-46, 2000 Jun 23.
Article in English | MEDLINE | ID: mdl-10854586

ABSTRACT

In the nervous system, signals transmitted across synapses are known to regulate gene expression in the postsynaptic cells. This process often involves membrane depolarization and subsequent elevation of intracellular Ca(2+). We have previously demonstrated in fetal cerebrocortical cells, that somatostatin (SS) mRNA levels can be induced by depolarizing agents such as high potassium concentrations and veratridine (VTD), and that these effects are calcium dependent. SS expression is regulated by cAMP, and in the cerebral cortex adenylate cyclase activity is regulated through fluctuations in intracellular Ca(2+) concentrations. The present experiments were undertaken to determine the mechanism by which calcium upregulates the levels of SS mRNA. Cerebrocortical cells from 17-day-old fetuses were exposed to the different agents for 24 h and the levels of SS mRNA were measured by Northern blot. Incubation of cells with the calcium channel antagonist nifedipine (Nf), the calcium chelating agent EGTA, calcium free KRB and the calcium calmodulin inhibitors trifluoroperazine (TFP) and the napthelene sulfonamide, W7, resulted in the inhibition of K(+)-induced SS mRNA levels. K(+)-evoked depolarization increased the intracellular concentration of cAMP and this effect was antagonized by verapamil (VPM). Forskolin (Fk) provoked a higher increment in cAMP concentration than potassium, although the induction of SS mRNA was more evident following K(+) depolarization indicating a lack of correlation between levels of cAMP and induction of SS mRNA. The role of K(+)-induced cAMP on the increment of SS mRNA that occurred upon membrane depolarization was further explored with the inhibitor of protein kinase A (PKA), Rp cAMP whose presence significantly reduced depolarization-induced SS mRNA levels. This study confirms that Ca(2+) influx is required for K(+)depolarization-induced stimulation of cAMP whereby the increment of SS mRNA is partly produced.


Subject(s)
Calcium Signaling/physiology , Cell Membrane/physiology , Cyclic AMP/metabolism , Diencephalon/metabolism , Neurons/metabolism , Potassium/metabolism , Somatostatin/genetics , Animals , Calcium/metabolism , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/metabolism , Cell Membrane/drug effects , Cells, Cultured , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Diencephalon/cytology , Diencephalon/drug effects , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Female , Fetus , Intracellular Fluid/metabolism , Neurons/cytology , Neurons/drug effects , Nifedipine/pharmacology , Pregnancy , RNA, Messenger/metabolism , Rats , Rats, Wistar , Sulfonamides/pharmacology , Thionucleotides/pharmacology , Trifluoperazine/pharmacology
9.
J Biol Chem ; 275(28): 21653-60, 2000 Jul 14.
Article in English | MEDLINE | ID: mdl-10801832

ABSTRACT

The mechanisms that control the emergence of different anterior pituitary cells from a common stem cell population are largely unknown. The immortalized GHFT cells derived from targeted expression of SV40 T antigen to mouse pituitary display characteristics of somatolactotropic progenitors in that they express the transcription factor GHF-1 (Pit-1) but not growth hormone (GH) or prolactin (PRL). We searched for factors capable of inducing lactotropic differentiation of GHFT cells. PRL gene expression was not observed in cells subjected to a variety of stimuli, which induce PRL gene expression in mature lactotropes. Only fibroblast growth factor-2 (FGF-2) was able to initiate the transcription, synthesis, and release of PRL in GHFT cells. However, induction of PRL expression was incomplete in FGF-2-treated cells, suggesting that additional factors are necessary to attain high levels of PRL transcription in fully differentiated lactotropes. We also show that the FGF-2 response element is located in the proximal PRL promoter. Stimulation of PRL expression by FGF-2 requires endogenous Ets factors and these factors as well as GHF-1 are expressed at low levels in the committed precursor, suggesting that these low levels are limiting for full PRL expression. Moreover, FGF-2 effect on lactotrope differentiation is mediated, at least partially, by stimulation of the Ras-signaling pathway. Our results suggest that, indeed, GHFT cells represent a valid model for studying lactotropic differentiation and that FGF-2 could play a key role both in initiating lactotrope differentiation and maintaining PRL expression.


Subject(s)
Cell Differentiation/physiology , Pituitary Gland, Anterior/drug effects , Prolactin/genetics , Transcription, Genetic/drug effects , Animals , Antigens, Polyomavirus Transforming/genetics , Cell Differentiation/drug effects , Cell Line, Transformed , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation/drug effects , Kinetics , Mice , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/physiology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ets , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Simian virus 40/genetics , Stem Cells/cytology , Transcription Factor Pit-1 , Transcription Factors/genetics , Transcription Factors/metabolism
10.
Mol Endocrinol ; 13(7): 1141-54, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10406465

ABSTRACT

PRL gene expression is dependent on the presence of the pituitary-specific transcription factor GHF-1/Pit-1, which is transcribed in a highly restricted manner in cells of the anterior pituitary. In pituitary GH3 cells, vitamin D increases the levels of PRL transcripts and stimulates the PRL promoter. We have analyzed the role of GHF-1 and of the vitamin D receptor (VDR) to confer vitamin D responsiveness to the PRL promoter. For this purpose we have used nonpituitary HeLa cells, which do not express GHF-1. We found that VDR activates the PRL promoter both in a ligand-dependent and -independent manner through a sequence located between positions -45/-27 in the proximal 5'-flanking region. This sequence also confers VDR and vitamin D responsiveness to a heterologous promoter. In the context of the PRL gene, VDR requires the presence of GHF-1 to activate the promoter. Truncation of the last 12 C-terminal amino acids of VDR, which contain the ligand-dependent activation function (AF2), abolishes regulation by vitamin D, suggesting that binding of coactivators to this region mediates ligand-dependent stimulation of the PRL promoter by the receptor. Indeed, expression of the coactivators, steroid hormone receptor coactivator-1 (SRC-1) and CREB-binding protein (CBP), significantly enhances the stimulatory effect of vitamin D mediated by the wild-type VDR but not by the AF2 mutant receptor. Furthermore, CBP also increases the activation of the PRL promoter by GHF-1 and the ligand-independent activation by both wild-type and mutant VDR.


Subject(s)
DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Prolactin/genetics , Receptors, Calcitriol/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Base Sequence , CREB-Binding Protein , DNA-Binding Proteins/genetics , Dimerization , Furylfuramide/metabolism , HeLa Cells/metabolism , Histone Acetyltransferases , Humans , Molecular Sequence Data , Mutation , Nuclear Receptor Coactivator 1 , Pituitary Neoplasms/genetics , Pituitary Neoplasms/metabolism , Prolactin/metabolism , Promoter Regions, Genetic , Rats , Receptors, Calcitriol/genetics , Receptors, Retinoic Acid/chemistry , Receptors, Retinoic Acid/metabolism , Response Elements/genetics , Retinoid X Receptors , Transcription Factor Pit-1 , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription, Genetic , Transcriptional Activation , Transfection , Tumor Cells, Cultured , Vitamin D/metabolism
11.
J Biol Chem ; 273(41): 26652-61, 1998 Oct 09.
Article in English | MEDLINE | ID: mdl-9756906

ABSTRACT

Although the effects of the peroxisome proliferator-activated receptors (PPARs) have been studied primarily in adipocytes and liver, the wide distribution of these receptors suggests that they might also play a role in other cell types. We present evidence that PPAR activators stimulate the expression of the prolactin gene in pituitary GH4C1 cells. Transfection assays in non-pituitary HeLa cells showed that stimulation of the prolactin promoter by PPARalpha requires the presence of the transcription factor GHF-1 (or Pit-1). Proximal promoter sequences confer responsiveness to PPARalpha, and activation by this receptor is lost concomitantly with the response to GHF-1. Surprisingly, expression of the retinoid X receptor (RXR) abolishes stimulation by PPARalpha. Furthermore, the promoter region that confers PPARalpha responsiveness does not contain a PPAR response element. This suggests that the transcriptional effect of PPARalpha might be mediated by protein-protein interactions rather than by binding of PPAR/RXR to the promoter. A direct interaction between PPARalpha and GHF-1 was confirmed by in vitro binding studies. Expression of the coactivators SRC-1 and CREB-binding protein, which bind to PPAR, also enhanced the responsiveness of the prolactin promoter to PPARalpha. Furthermore, CREB-binding protein also significantly increased activation by GHF-1, and both proteins associated in vitro. Thus, PPARalpha, a receptor that normally acts as a ligand-dependent transcription factor by binding to specific DNA sequences in one context, can also stimulate the prolactin promoter by association with GHF-1 and coactivator proteins.


Subject(s)
Gene Expression Regulation/physiology , Pituitary Gland/metabolism , Prolactin/genetics , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/physiology , Base Sequence , Binding Sites , Cell Line , Cell Nucleus/metabolism , Chloramphenicol O-Acetyltransferase/genetics , DNA Primers , HeLa Cells , Humans , Nuclear Proteins/metabolism , Pituitary Gland/cytology , Promoter Regions, Genetic , Protein Binding , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Estrogen/physiology , Receptors, Retinoic Acid/physiology , Retinoid X Receptors , Trans-Activators/metabolism , Transcription Factors/metabolism
12.
Cell Calcium ; 23(5): 323-38, 1998 May.
Article in English | MEDLINE | ID: mdl-9681195

ABSTRACT

Human calmodulin is encoded by three genes CALM1, CALM2 and CALM3 located on different chromosomes. To complete the characterization of this family, the exon-intron structure of CALM2 was solved by a combination of genomic DNA library screening and genomic PCR amplification. Intron interruptions were found at identical positions in human CALM2 as in CALM1 and CALM3; however, the overall size of CALM2 (16 kb) was almost twice that of the other two human CALM genes. Over 1 kb of the 5' flanking sequence of human CALM2 were determined, revealing the presence of a TATA-like sequence 27 nucleotides upstream of the transcriptional start site and several conserved sequence elements possibly involved in the regulation of this gene. To determine if differential transcriptional activity plays a major role in regulating cellular calmodulin levels, we directly measured and compared the mRNA abundance and transcriptional activity of the three CALM genes in proliferating human teratoma cells. CALM3 was at least 5-fold more actively transcribed than CALM1 or CALM2. CALM transcriptional activity agreed well with the mRNA abundance profile in the teratoma cells. In transient transfections using luciferase reporter genes driven by 1 kb of the 5' flanking DNA of the three CALM genes, the promoter activity correlated with the endogenous CALM transcriptional activity, but only when the 5' untranslated regions were included in the constructs. We conclude that the CALM gene family is differentially active at the transcriptional level in teratoma cells and that the 5' untranslated regions are necessary to recover full promoter activation.


Subject(s)
Calmodulin/genetics , Genes , Transcription, Genetic , Amino Acid Sequence , Base Sequence , Chloramphenicol O-Acetyltransferase/biosynthesis , Cloning, Molecular , DNA, Complementary/genetics , Exons/genetics , Gene Expression Regulation , Gene Library , Genes, Reporter , Humans , Introns/genetics , Molecular Sequence Data , Multigene Family , Organ Specificity , Polymerase Chain Reaction , RNA Splicing , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Teratocarcinoma/pathology , Transfection , Tumor Cells, Cultured
13.
Oncogene ; 16(15): 1981-91, 1998 Apr 16.
Article in English | MEDLINE | ID: mdl-9591782

ABSTRACT

We have examined the influence of insulin-like growth factor I (IGF-1) on prolactin gene expression in rat pituitary GH4C1 cells. Incubation with IGF-1 increases prolactin mRNA levels and activates the prolactin promoter in transient transfection assays. A similar degree of activation is observed with constructs extending to -3000 and -176 base pairs of the prolactin 5' flanking region, indicating that the IGF-1 response element is located in the proximal promoter sequences. A plasmid containing 101 base pairs shows a partial stimulation by IGF-1, and the response is lost in a deletion to -76 base pairs. The Ras oncoproteins have been implicated as a critical signaling component in mediating the effect of growth factor receptor tyrosine kinases. Expression of oncogenic RasVal12 mimics the effect of IGF-1 on the prolactin promoter, and a dominant negative Ras, RasAsn17, blocks IGF-1-mediated stimulation. Dominant negative mitogen-activated protein kinase (MAPK) also reduces the activation of the prolactin promoter by IGF-1. Ets transcription factors have been described to lie downstream of Ras and MAPK in the signaling pathway leading to prolactin gene activation. Mutation of two Ets binding sites in the promoter region between -101 and -76 abolishes the response to IGF-1. Furthermore, a dominant negative Ets vector strongly reduces the response of the prolactin promoter to IGF-1 and Ras. The endogenous concentration of Ets-related proteins is not limiting in GH4C1 cells for the IGF-1 effect. However, c-Ets-1 and GHF-1 act synergistically in HeLa cells with the IGF-1 receptor, reconstituting pituitary IGF-1 responsiveness. The response to IGF-1 in GH4C1 cells is still observed after transfection with RasVal12 suggesting that, although Ras is required, IGF-1 could stimulate other pathway/s in addition to Ras. Wortmanin, an inhibitor of phosphatidylinositol-3 kinase (PI-3 kinase), also prevents the response of the prolactin promoter to IGF-1. These results show that both the Ras/MAPK/Ets pathway, as well as the activation of PI-3 kinase are involved in the signaling mechanism leading to prolactin expression by IGF-1 in GH4C1 cells.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Phosphatidylinositol 3-Kinases/physiology , Prolactin/genetics , Proto-Oncogene Proteins/physiology , Transcription Factors/physiology , ras Proteins/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Cyclic AMP/physiology , Gene Expression/drug effects , Promoter Regions, Genetic , Proto-Oncogene Proteins c-ets , RNA, Messenger/analysis , Rats , Tumor Cells, Cultured
14.
Endocrinology ; 137(10): 4384-91, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8828499

ABSTRACT

The neuropeptide somatostatin (SS) plays a role as a modulator of cognitive functions and as a potential tropic factor in the central nervous system. A reduction in SS levels has been demonstrated in the aging brain and in dementia. In addition, insulin-like growth factor I (IGF-I) acts as a paracrine factor in multiple GH actions and is also found in the cerebral hemispheres, where it exerts neurotropic effects. We used aging rats as an in vivo model of GH deficiency to study the possible participation of exogenous GH in the modulation of the cerebral hemispheric SS and IGF-I. Two sets of experiments were carried out. In the first set, the age-related patterns of GH, IGF-I, and SS in the serum, pituitary, and cerebral hemispheres were established. In the second experimental set, 90-day-old (adult) and 2-yr-old (aging) male rats received recombinant human GH (200 micrograms/ sc-day) or vehicle for 7 consecutive days. The serum levels of rat GH and IGF-I as well as pituitary GH messenger RNA decreased in 2-yr-old rats compared with those in adult rats. After GH treatment, pituitary GH messenger RNA levels decreased markedly in the 90-day-old and 2-yr-old rats. Serum immunoreactive GH decreased in the adult animals, whereas it remained unaffected in the aging ones, whereas serum IGF-I levels were not altered by GH treatment in either group. Immunoreactive levels and messenger RNA of both SS and IGF-I were low in the cerebral hemispheres of aging rats, but were restored to the levels found in adult rats after GH treatment. As treatment did not induce changes in the serum IGF-I levels, these results provide evidence of a stimulatory action of peripherally administered GH on the regulation of SS and IGF-I genes in the aging rat in the central nervous system. These data also show a new target action for GH and could provide a molecular basis for the improvement of some symptoms of GH deficiency that occurs after recombinant human GH treatment.


Subject(s)
Aging/physiology , Brain/drug effects , Gene Expression/drug effects , Human Growth Hormone/pharmacology , Insulin-Like Growth Factor I/genetics , Somatostatin/genetics , Aging/blood , Animals , Brain/physiology , Human Growth Hormone/blood , Humans , Insulin-Like Growth Factor I/analysis , Male , Rats , Rats, Wistar , Receptors, Somatotropin/genetics , Recombinant Proteins
15.
Brain Res Mol Brain Res ; 35(1-2): 103-10, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8717345

ABSTRACT

The stimulatory effect of veratridine (VTD) depolarization upon somatostatin mRNA (SS mRNA) levels in primary cultures of fetal cerebrocortical cells was analyzed. Depolarizing stimuli, such as 100 microM VTD exposure for 30 min, elicited an increase in immunoreactive somatostatin (IR-SS) release to the media without affecting SS mRNA levels. These levels increased when exposure to depolarization stimuli was prolonged up to 3 or more hours. At this time, veratridine acted as a secretagogue, stimulating somatostatin secretion, but was also effective in stimulating somatostatin mRNA levels. These changes were blunted by the Na+ channel blockade tetrodotoxin (TTX), and partially abolished by the Ca2+ channel antagonist, verapamil (VPM). To study whether VTD may affect mRNA stability we determine the rate of disappearance of SS mRNA after inhibition of transcription by actinomycin D and demonstrated that VTD stimulation did not stabilize the SS mRNA. These results indicate that the induction of SS mRNA expression by VTD involves the modulation of Ca2+ and Na+ channels. The time course study confirmed that the VTD-induced SS mRNA accumulation is time-dependent, and requires a prolonged exposure to stimulate SS gene expression. VTD stimulation does not modify the SS mRNA rate of degradation.


Subject(s)
Cerebral Cortex/physiology , Gene Expression Regulation/drug effects , Neurons/physiology , Somatostatin/biosynthesis , Veratridine/pharmacology , Animals , Cells, Cultured , Cerebral Cortex/drug effects , Dactinomycin/pharmacology , Female , Fetus , Kinetics , Membrane Potentials/drug effects , Neurons/cytology , Neurons/drug effects , Pregnancy , Rats , Rats, Wistar , Somatostatin/metabolism , Tetrodotoxin/pharmacology , Verapamil/pharmacology
16.
Neuroendocrinology ; 61(1): 31-5, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7731495

ABSTRACT

It is well known that chronic supraphysiological doses of glucocorticoids (GC) inhibit GH secretion in vivo, and stimulate GH secretion from the somatotropes in vitro. It has been suggested that GC exert an inhibitory role in the hypothalamus surpassing the GC-positive effect at the somatotrope level. To test the hypothesis that GC can affect growth hormone-releasing releasing factor (GRF) and somatostatin (SS) at the hypothalamic level, we studied the effect of corticosterone on the immunoreactive content of GRF (IR-GRF) and SS (IR-SS) in cells and media of fetal hypothalamic cells in culture. After 20 days in culture, cells were incubated with serum-free medium containing corticosterone (from 0.3 to 300 nM) for 48 h. Corticosterone had a dual effect on IR-GRF. Concentrations in the range of the glucocorticoid receptor Kd (3 nM) increased peptide content, whereas higher concentrations (30 and 300 nM) decreased IR-GRF content in cells and media. Conversely, corticosterone increased SS cell content, only at a concentration of 3 nM, inducing a 2- to 3-fold increment in media content with the highest doses (30 and 300 nM). These results demonstrated that both GRF and SS are modulated by corticosterone in primary fetal rat hypothalamic cultures. Whereas GRF exhibited a dual response, stimulatory and inhibitory, at low and high corticosterone doses, respectively, SS showed a parallel increase with the corticosterone concentrations.


Subject(s)
Corticosterone/pharmacology , Fetal Proteins/analysis , Growth Hormone-Releasing Hormone/analysis , Hypothalamus/drug effects , Somatostatin/analysis , Animals , Cells, Cultured , Hypothalamus/chemistry , Hypothalamus/embryology , Radioimmunoassay , Rats , Rats, Wistar
17.
Peptides ; 16(2): 249-53, 1995.
Article in English | MEDLINE | ID: mdl-7784255

ABSTRACT

The effect of triiodothyronine (T3) on somatostatin (SS) mRNA levels in cultured fetal rat cerebrocortical cells was studied. Two different experimental approaches were sought. They differed in the length of time in which cells were deprived of thyroid hormones prior to the addition of exogenous T3. When the cells were not deprived of thyroid hormones, T3 caused a dose-related decrease in SS mRNA content at all doses tested. However, when the cells were deprived of T3 for 24 h, a biphasic effect was observed. These findings suggest that T3 regulates SS gene expression in fetal cultured cerebrocortical cells.


Subject(s)
Cerebral Cortex/metabolism , Gene Expression/drug effects , Neurons/metabolism , Somatostatin/biosynthesis , Triiodothyronine/pharmacology , Animals , Blotting, Northern , Cells, Cultured , Dose-Response Relationship, Drug , Fetus , Kinetics , Neurons/drug effects , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Rats , Time Factors
18.
Endocrinology ; 135(6): 2526-32, 1994 Dec.
Article in English | MEDLINE | ID: mdl-7988440

ABSTRACT

The effects of recombinant human insulin-like growth factor I (IGF-I) on both vasoactive intestinal peptide (VIP) and PRL production and gene expression were studied using rat anterior pituitary cell cultures grown in serum-free defined medium. We also examined whether pituitary VIP could be involved in the PRL response to IGF-I and hence in a paracrine regulatory system. Exposure of cultured anterior pituitary cells to IGF-I (2.6 nM) for 3 h caused a significant decrease in both VIP content and media PRL. Treatment with IGF-I (from 0.65-5.2 nM) for 48 h increased VIP production and VIP messenger RNA (mRNA) accumulation, whereas only an increase in media and intracellular PRL content without changes in mRNA was observed. In all these experiments, IGF-I led to a decrease in both GH secretion and expression. Immunoglobulins G purified from VIP antiserum inhibited IGF-I-induced PRL release without affecting intracellular and mRNA levels. The inhibition of both GH secretion and gene expression induced by IGF-I was not blocked by VIP antiserum. In conclusion, these results indicate that IGF-I induces VIP gene expression, and its secretion and also increases PRL secretion. The effect of IGF-I on PRL release is specifically mediated by VIP through a paracrine or autocrine mechanism.


Subject(s)
Gene Expression/drug effects , Insulin-Like Growth Factor I/pharmacology , Pituitary Gland, Anterior/drug effects , Prolactin/metabolism , Vasoactive Intestinal Peptide/genetics , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Male , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/physiopathology , Prolactin/biosynthesis , Rats , Rats, Wistar , Time Factors , Vasoactive Intestinal Peptide/biosynthesis , Vasoactive Intestinal Peptide/physiology
19.
J Neurosci ; 14(3 Pt 1): 1053-9, 1994 Mar.
Article in English | MEDLINE | ID: mdl-7907136

ABSTRACT

The stimulatory effect of potassium depolarization upon somatostatin (SS) mRNA levels in primary cultures of fetal cerebrocortical cells was analyzed. Depolarizing stimuli, such as 56 mM K+ exposure for 30 min, elicited an increase in immunoreactive somatostatin (IR-SS) release to the media and decreased SS mRNA levels. These were increased when exposure to depolarization stimuli was prolonged up to 3 or more hr. At this time, potassium (30 and 56 mM) acted as a secretagogue, stimulating SS secretion, but was also effective in stimulating SS mRNA levels, suggesting that SS secretion can be coupled to SS mRNA accumulation. These changes were inhibited by the Ca2+ channel antagonist verapamil. In contrast, Na+ channel blockade by TTX did not modify the 24 hr potassium-induced increase in SS mRNA, although it partially abolished potassium-induced SS secretion. Examination of the rate of disappearance of SS mRNA levels after inhibition of mRNA transcription by actinomycin-D revealed that K+ stimulation of cerebrocortical cells stabilized the SS mRNA. These results suggest that the induction of SS mRNA expression by K+ is dose dependent, and involves the modulation of ion channels. The time-course study confirmed that the K(+)-induced SS mRNA accumulation is time dependent, chronic activation of the Ca2+ channels being necessary to stimulate SS gene expression. K+ stimulation may also increase the level of SS mRNA in cerebrocortical cells by reducing its rate of degradation.


Subject(s)
Cerebral Cortex/physiology , Gene Expression , Potassium/metabolism , Somatostatin/genetics , Animals , Cells, Cultured , Cerebral Cortex/metabolism , Fetus/physiology , RNA, Messenger/metabolism , Rats , Tetrodotoxin/pharmacology , Verapamil/pharmacology
20.
Neuroendocrinology ; 58(6): 655-65, 1993 Dec.
Article in English | MEDLINE | ID: mdl-7907399

ABSTRACT

Information about growth hormone-releasing factor (GRF) regulation by somatostatin, GH and IGF-I is scarce and controversial. This could be due to the in vivo interactions among these signals and the lack of models for individualizing the action of one of them from the others upon GRF regulation. The aim of the present work was to study GRF regulation by these signals, using primary fetal rat hypothalamic-brain stem cell cocultures. Coculturing of these two cytotypes increases hypothalamic immunoreactive rat GRF (IR-rGRF) content in cells by 45% and in media by 36%. The effect of SS on GRF in cocultures was examined by using a multiple approach: (1) depleting endogenous SS by adding 1 mM cysteamine (CSH); (2) blocking endogenous SS by incubation with SS antiserum, and (3) incubating with synthetic SS14 at different concentrations and exposure periods. 1 mM CSH depleted IR-SS content (pg/plate, mean +/- SE) in cells (CSH-treated: 68 +/- 8 vs. control: 322 +/- 10, p < 0.01) and media (CSH-treated: 211 +/- 15 vs. control: 880 +/- 70; p < 0.01). In the CSH-induced SS-depleted cultures, a slight reduction in the IR-rGRF content in cells was observed (CSH-treated: 93.5 +/- 4.5 vs. control: 111 +/- 6; p < 0.05), with no effect on media content. When SS antiserum was added to plates, there was a slight reduction in the IR-rGRF content in cells and media, but it was not significantly different from the controls. However, SS14 (10(-10)-10(-8) M) could not modify IR-rGRF content in media and cells. The GH effect on IR-rGRF was studied in the absence of CSH and in CSH-induced SS-depleted cultures. GH (5 microM, 24 h) decreased (52%) the IR-rGRF content in media (GH-treated: 28.7 +/- 4.6 vs. control: 60.2 +/- 7; p < 0.01) without causing changes in cell content. In SS-depleted cultures, the inhibitory action of GH on media IR-rGRF was greater (62% decrease) (GH-treated: not detected, control 56 +/- 10; p < 0.01) and also affected IR-rGRF cell content (GH-treated: 64.3 +/- 7.3 vs. control: 160 +/- 9.6; p < 0.01). In the same experiments, GH increased IR-SS content in cells (GH-treated: 31.8 +/- 4.6 vs. control 20.9 +/- 0.5; p < 0.01) and in media (GH-treated: 413 +/- 7 vs. control: 286 +/- 9; p < 0.01). 1 mM CSH again depleted IR-SS content and abolished the GH stimulatory effect.(ABSTRACT TRUNCATED AT 400 WORDS)


Subject(s)
Brain Stem/metabolism , Growth Hormone-Releasing Hormone/metabolism , Growth Hormone/pharmacology , Hypothalamus/metabolism , Insulin-Like Growth Factor I/pharmacology , Somatostatin/pharmacology , Animals , Brain Stem/drug effects , Cells, Cultured , Culture Media , Growth Hormone/immunology , Hypothalamus/drug effects , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Rats , Rats, Wistar , Recombinant Proteins/pharmacology , Somatostatin/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...