Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 119
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Regul Homeost Agents ; 27(1): 143-54, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23489694

RESUMEN

Glioblastoma multiforme (GBM) is among the most devastating human tumors being rapidly fatal despite aggressive surgery, radiation and chemotherapies. It is characterized by extensive dissemination of tumor cells within the brain that hinders complete surgical resection. GBM tumor initiating-cells (TICs) are a rare subpopulation of cells responsible for tumor development, growth, invasiveness and recurrence after chemotherapy. TICs from human GBM can be selected in vitro using the same conditions permissive for the growth of normal neural cells, of which share some features including marker expression, self-renewal capacity, long-term proliferation, and ability to differentiate into neuronal and glial cells. EGFR overexpression and its constitutive activation is one of the most important signaling alteration identified in GBM, and its pharmacological targeting represents an attractive therapeutic goal. We previously demonstrated that human GBM TICs have different sensitivity to the EGFR kinase inhibitors erlotinib and gefitinib, depending on the differential modulation of downstream signaling cascades. In this work we investigated the mechanisms of resistance to erlotinib in two human GBM TIC cultures, analyzing EGF and bFGF individual contribution to proliferation, clonogenicity, and migration. We demonstrated the presence of a small cell subpopulation whose proliferation is supported by EGF and a larger one mainly dependent on bFGF. Thus, insensitivity to EGFR kinase inhibitors as far as TIC proliferation results from a predominant FGFR activation that hides the inhibitory effects induced on EGFR signaling. Conversely, EGF and bFGF induced cell migration with similar efficacy. In addition, unlike neural stem/progenitors cells, the removal of chondroitin sulphate proteoglycans from cell surface was unable to discern EGF- and bFGF-dependent subpopulations in GBM TICs.


Asunto(s)
Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/antagonistas & inhibidores , Factor 2 de Crecimiento de Fibroblastos/farmacología , Glioblastoma/patología , Células Madre Neoplásicas/patología , Inhibidores de Proteínas Quinasas/farmacología , Anciano , Recuento de Células , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Sulfatos de Condroitina/metabolismo , Células Clonales , Ensayos de Selección de Medicamentos Antitumorales , Receptores ErbB/metabolismo , Glioblastoma/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/patología , Factores de Tiempo , Ensayo de Tumor de Célula Madre
2.
J Dermatolog Treat ; 33(1): 401-407, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32349568

RESUMEN

BACKGROUND: Cyclosporine A (CsA) is one of the systemic therapeutic options for moderate-to-severe psoriasis, based on its efficacy and rapidity of action. The current study investigated the response to CsA in patients with moderate-to-severe plaque psoriasis. MATERIALS AND METHODS: TRANSITION was an observational, cross-sectional, multicentre study which evaluated the proportion of partial- and suboptimal-responders among patients with moderate-to-severe plaque psoriasis treated with continuous CsA for ≥12 weeks. Patients demonstrating a Psoriasis Area and Severity Index (PASI) response of ≥90, ≥75 and <90, ≥50 and <75 and <50 were defined as responders, suboptimal-responders, partial-responders, and non-responders, respectively. RESULTS: A total of 196 patients (mean age, 46.6 years; 62.8% males) from 14 sites in Italy were evaluated. At the study visit, the mean (SD) PASI score was 4.2(5.5) compared with 15.3(7.1) prior to the last CsA cycle. For response categories, 39.8%, 22.4%, 16.8%, and 20.9% of patients were responders, suboptimal-responders, partial-responders, and non-responders to CsA treatment. Overall, 28.6% of patients permanently discontinued treatment with CsA (lack of efficacy [10.2%], poor tolerability and voluntary discontinuation [3.6% each], and other [11.7%]). CONCLUSION: Patients were only partially satisfied with CsA treatment, reporting measurable impact on quality of life. Only 40% patients showed a satisfactory response to CsA.


Asunto(s)
Ciclosporina , Psoriasis , Estudios Transversales , Ciclosporina/uso terapéutico , Femenino , Humanos , Masculino , Persona de Mediana Edad , Psoriasis/tratamiento farmacológico , Calidad de Vida , Índice de Severidad de la Enfermedad , Resultado del Tratamiento
3.
J Biol Regul Homeost Agents ; 24(1): 27-39, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20385069

RESUMEN

In this paper we analyzed the determinants and the structural effects of the interaction of human prion protein fragment 90-231 (HuPrP) with humic substances, (HS) including humic (HA) and fulvic (FA) acids, natural refractory organic polyanions widely diffused in soils and waters. We show that this interaction is mainly driven by non-specific electrostatic attraction involving regions situated within alpha-helix A and beta-sheet S1 of human PrP. FA binding to HuPrP altered its ability to acquire some PrPSc-like characteristics induced by the mild thermal denaturation of the peptide (1 h at 53 degrees C). In particular, in the presence of FA, HuPrP shows a reduced amount of beta-sheet content (as demonstrated by the reduced binding of thioflavin T), an increased sensitivity to protease K and an inhibition of the entering in the fibrillogenic pathway. FA/HuPrP interaction caused the aggregation of the peptide in unstructured macrocomplexes, as demonstrated by the altered electrophoretic migration in semi-denaturing detergent-agarose gel assay. Importantly, in the presence of FA the rate of internalization of HuPrP in human neuroblastoma cells was significantly reduced as compared to that of the beta-structured peptide. Therefore, HS inhibited the acquisition of PrP(Sc)-like structural properties that, in turn, are responsible for HuPrP intracellular accumulation and lead to neuronal death. Important implications of these data are that HuPrP-HS complexes, being unable to be internalized in living cells may represent a molecular mechanism for the reduced transmission of prion transmission from HS-rich soil also in the presence of contamination from infected animals.


Asunto(s)
Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Priones/química , Priones/metabolismo , Secuencia de Aminoácidos , Benzopiranos/metabolismo , Benzopiranos/farmacología , Benzotiazoles , Línea Celular , Endopeptidasa K/metabolismo , Humanos , Sustancias Húmicas , Técnicas In Vitro , Modelos Moleculares , Datos de Secuencia Molecular , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/toxicidad , Proteínas PrPC/química , Proteínas PrPC/metabolismo , Proteínas PrPSc/química , Proteínas PrPSc/metabolismo , Priones/genética , Priones/toxicidad , Unión Proteica , Estructura Secundaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Suelo/análisis , Espectrometría de Fluorescencia , Electricidad Estática , Tiazoles/metabolismo
4.
G Ital Dermatol Venereol ; 145(3): 319-22, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20461041

RESUMEN

AIM AND METHODS: The treatment of mild-moderate acne with topical drugs in association with appropriate cosmetics is currently the golden standard. The tolerability and efficacy of a cream formulated with a new mix of alpha-hydroxy acids (Hyseac AHA cream) in 248 patients with mild-moderate acne (comedonic, inflammatory, or mixed) have been investigated in a multicenter, non-randomized, open study by 10 dermatologists from different Italian areas during their routine practice. The medication with Hyseac AHA cream was prescribed at first consultation for 60 days, twice a day, either as a monotherapy (49.2% patients) or in association with a pharmacological treatment (50.2%). RESULTS: The tolerability was good to excellent in 92.3% patients, without significant differences between patients using AHA cream in monotherapy (90.0%) or associated with concomitant pharmacological treatment (97.6%). The efficacy was overall high in 64.2% patients, again without significant differences related to concomitant pharmacological treatment or not (64.8% vs. 63.3%) and/or the acne clinical type (comedonic vs. inflammatory vs. mixed: 69.2% vs. 66.7% vs. 58%). CONCLUSION: The results of this study, while confirming the high tolerability and efficacy of this AHA cream in the treatment of mild/moderate acne, reasonably suggest its possible use also in monotherapy. Furthermore, its use can be reasonably hypothesized as a maintenance treatment after specific pharmacological treatment even in more severe acne types.


Asunto(s)
Acné Vulgar/tratamiento farmacológico , Hidroxiácidos/uso terapéutico , Adolescente , Adulto , Niño , Femenino , Humanos , Masculino , Persona de Mediana Edad , Índice de Severidad de la Enfermedad , Adulto Joven
5.
Science ; 256(5060): 1215-7, 1992 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-1350382

RESUMEN

The growth-inhibiting peptide hormone somatostatin stimulates phosphotyrosine phosphatase activity in the human pancreatic cell line MIA PaCa-2. This hormonal activation was mediated by a pertussis toxin-sensitive guanosine 5'-triphosphate-binding protein (G protein) in the membranes of these cells. Activation of this G protein by somatostatin stimulated the dephosphorylation of exogenous epidermal growth factor receptor prepared from A-431 cells in vitro. This pathway may mediate the antineoplastic action of somatostatin in these cells and in human tumors and could represent a general mechanism of G protein coupling that is utilized by normal cells in the hormonal control of cell growth.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Somatostatina/farmacología , Adenosina Trifosfato/metabolismo , Carcinoma de Células Escamosas , Línea Celular , Membrana Celular/metabolismo , Relación Dosis-Respuesta a Droga , Activación Enzimática , Receptores ErbB/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/farmacología , Guanosina Difosfato/análogos & derivados , Guanosina Difosfato/farmacología , Humanos , Cinética , Neoplasias Pancreáticas , Péptidos/metabolismo , Toxina del Pertussis , Fosforilación , Proteínas Quinasas/metabolismo , Tionucleótidos/farmacología , Factores de Virulencia de Bordetella/farmacología
6.
Br J Pharmacol ; 155(2): 198-209, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18587421

RESUMEN

BACKGROUND AND PURPOSE: Cyclooxygenase-2 (COX-2) is expressed in colonic neoplasms, where it supports cell proliferation via prostaglandin E(2) (PGE(2)) production. This study investigated the effects of somatostatin-14 on COX-2 expression, PGE(2) production and proliferation in colon cancer cells. EXPERIMENTAL APPROACH: Human colon adenocarcinoma cell lines Caco-2, HT-29 and HCT116 were used. The following techniques were employed: colourimetric assay for cell growth; 5-bromo-2'-deoxyuridine assay for DNA synthesis; enzyme immunoassay for PGE(2); COX-2 mRNA silencing; RT-PCR or Western blot for somatostatin receptor subtypes, cyclooxygenase isoforms, phosphorylated-ERK-1/ERK-2 and phosphorylated-Akt. KEY RESULTS: HT-29 and Caco-2 cells expressed COX-2 and somatostatin receptors (sst(3/4/5) and sst(3/5), respectively). HCT116 cells did express somatostatin receptors (sst(2/3/5)), but not COX-2. Somatostatin-14 inhibited basal COX-2 expression, PGE(2) production, DNA synthesis and growth in Caco-2 cells and these effects were prevented by BN81658 (sst(3) receptor antagonist). Basal proliferation of HT-29, HCT116 and COX-2-silenced Caco-2 cells was not affected by somatostatin-14. Stimulation of HT-29 cells with gastrin-17 elicited increments of ERK-1/ERK-2 and Akt phosphorylation, COX-2 expression, PGE(2) production, DNA synthesis and cell growth, which were all counteracted by somatostatin-14. Somatostatin-14-induced inhibition of COX-2 expression, PGE(2) production and DNA synthesis were blocked by BIM23056 (sst(5) receptor antagonist). CONCLUSIONS AND IMPLICATIONS: Somatostatin decreases COX-2 expression and function in colon cancer cells via activation of sst(3) or sst(5) receptors, and these effects contribute to the inhibitory action of somatostatin on cell proliferation. These findings can be relevant to the development of therapeutic strategies based on the modulation of the COX-2 pathway.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/patología , Inhibidores de la Ciclooxigenasa 2/farmacología , Ciclooxigenasa 2/metabolismo , Somatostatina/farmacología , Células CACO-2 , Colon/patología , Ciclooxigenasa 2/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Gastrinas/metabolismo , Células HT29 , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Oligopéptidos/metabolismo , Proteína Quinasa C/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo
7.
Science ; 361(6403)2018 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-30115782

RESUMEN

The coordinated expression of highly related homoeologous genes in polyploid species underlies the phenotypes of many of the world's major crops. Here we combine extensive gene expression datasets to produce a comprehensive, genome-wide analysis of homoeolog expression patterns in hexaploid bread wheat. Bias in homoeolog expression varies between tissues, with ~30% of wheat homoeologs showing nonbalanced expression. We found expression asymmetries along wheat chromosomes, with homoeologs showing the largest inter-tissue, inter-cultivar, and coding sequence variation, most often located in high-recombination distal ends of chromosomes. These transcriptionally dynamic genes potentially represent the first steps toward neo- or subfunctionalization of wheat homoeologs. Coexpression networks reveal extensive coordination of homoeologs throughout development and, alongside a detailed expression atlas, provide a framework to target candidate genes underpinning agronomic traits in wheat.


Asunto(s)
Regulación de la Expresión Génica de las Plantas , Poliploidía , Transcripción Genética , Triticum/genética , Pan , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Genoma de Planta , ARN de Planta/genética , Análisis de Secuencia de ARN , Triticum/crecimiento & desarrollo
8.
Neuroscience ; 142(4): 991-7, 2006 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-16949212

RESUMEN

Hereditary creatine transporter deficiency causes brain damage, despite the brain having the enzymes to synthesize creatine. Such damage occurring despite an endogenous synthesis is not easily explained. This condition is incurable, because creatine may not be delivered to the brain without its transporter. Creatine-derived compounds that crossed the blood-brain barrier in a transporter-independent fashion would be useful in the therapy of hereditary creatine transporter deficiency, and possibly also in neuroprotection against brain anoxia or ischemia. We tested the double hypothesis that: (1) the creatine carrier is needed to make creatine cross the plasma membrane of brain cells and (2) creatine-derived molecules may cross this plasma membrane independently of the creatine carrier. In in vitro mouse hippocampal slices, incubation with creatine increased creatine and phosphocreatine content of the tissue. Inhibition of the creatine transporter with 3-guanidinopropionic acid (GPA) dose-dependently prevented this increase. Incubation with creatine benzyl ester (CrOBzl) or phosphocreatine-Mg-complex acetate (PCr-Mg-CPLX) increased tissue creatine content, not phosphocreatine. This increase was not prevented by GPA. Thus, the creatine transporter is required for creatine uptake through the plasma membrane. Since there is a strong indication that creatine in the brain is mainly synthesized by glial cells and transferred to neurons, this might explain why hereditary transporter deficiency is attended by severe brain damage despite the possibility of an endogenous synthesis. CrOBzl and PCr-Mg-CPLX cross the plasma membrane in a transporter-independent way, and might be useful in the therapy of hereditary creatine transporter deficiency. They may also prove useful in the therapy of brain anoxia or ischemia.


Asunto(s)
Encéfalo/metabolismo , Membrana Celular/metabolismo , Creatina/deficiencia , Proteínas de Transporte de Membrana/metabolismo , Neuronas/metabolismo , Animales , Transporte Biológico Activo/efectos de los fármacos , Transporte Biológico Activo/fisiología , Encéfalo/efectos de los fármacos , Encefalopatías Metabólicas/tratamiento farmacológico , Encefalopatías Metabólicas/metabolismo , Encefalopatías Metabólicas/fisiopatología , Membrana Celular/efectos de los fármacos , Creatina/análogos & derivados , Creatina/farmacología , Inhibidores Enzimáticos/farmacología , Guanidinas/farmacología , Masculino , Proteínas de Transporte de Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Neuronas/efectos de los fármacos , Técnicas de Cultivo de Órganos , Propionatos/farmacología
9.
Int J Immunopathol Pharmacol ; 19(2): 339-56, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16831301

RESUMEN

The transition of prion protein from a mainly alpha-structured isoform (PrPC) to a beta sheet-containing protein (PrPSc) represents a major pathogenetic mechanism in prion diseases. To study the role of PrP structural conformation in prion-dependent neurodegeneration, we analysed the neurotoxicity of PrP in alpha and beta conformations, using a recombinant protein encompassing amino acids 90-231 of the human PrP (hPrP90-231). Using controlled thermal denaturation (53 degrees C, 1h) we converted hPrP90-231 in a structural isoform displaying PrPSc-related characteristics: high beta sheet content, increased aggregability and a slight increase in the resistance to protease K. In virtue of these structural changes, hPrP90-231 powerfully affected the survival of SH-SY5Y cells, inducing a caspase-3 and p38- dependent apoptosis. Conversely, in the native alpha-helix-rich conformation, hPrP90-231 did not show significant cell toxicity. The relationship between the structural state of hPrP90-231 and its neurotoxicity was demonstrated, inducing the thermal denaturation of the peptide in the presence of Congo red that prevented both the transition of hPrP90-231 into a beta-rich isoform and the acquisition of toxic properties. In conclusion, we report that the toxicity of hPrP90-231 is dependent on its three-dimensional structure, as is supposed to occur for the pathogen PrP during TSE.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas PrPC/química , Proteínas PrPC/farmacología , Amiloide/biosíntesis , Benzotiazoles , Caspasa 3 , Caspasa 7 , Caspasas/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Dicroismo Circular , Endopeptidasa K/química , Colorantes Fluorescentes , Humanos , Hidrólisis , Immunoblotting , L-Lactato Deshidrogenasa/metabolismo , Microscopía Electrónica , Necrosis , Conformación Proteica , Desnaturalización Proteica , Estructura Secundaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacología , Relación Estructura-Actividad , Sales de Tetrazolio , Tiazoles/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
10.
Cell Signal ; 10(2): 75-84, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9481481

RESUMEN

During the last few years several studies have been undertaken to characterise the role of gp120, the HIV-1 envelope glycoprotein, in the pathogenesis of neurological defects associated with AIDS. However, neurons did not appear to be the main target of the virus, since the widespread neuronal damage is not associated with a productive viral infection in neurons. The current opinion supports the hypothesis that an indirect mechanism exists to explain the neuronal cell death which occurs in patients infected by HIV-1. In particular, several reports suggest that gp120 may be the main candidate as mediator of the neurological deficits during HIV-1 infection and demonstrate that this molecule affects neuronal survival through a direct interaction with non-neuronal cell types such as monocytes, macrophages/microglia and astrocytes.


Asunto(s)
Complejo SIDA Demencia/patología , Complejo SIDA Demencia/virología , Proteína gp120 de Envoltorio del VIH/fisiología , VIH-1/patogenicidad , Neuronas/patología , Transducción de Señal , Complejo SIDA Demencia/tratamiento farmacológico , Apoptosis , Humanos
11.
Int J Immunopathol Pharmacol ; 18(1): 95-112, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15698515

RESUMEN

Peptides corresponding to three alpha helices present in the C-terminal region of the human prion protein have been synthesized and their structural autonomy analyzed by circular dichroism (CD) and NMR spectroscopy. The results obtained indicate that the protein fragment corresponding to the alpha 3-helix, in contrast to alpha 1 and alpha 2 peptides, shows a complete structural autonomy. The chemical shifts values found for NH and CHalpha resonance of the isolated alpha 3 peptide, formed by 30 aminoacid residues, were markedly and surprisingly similar to the corresponding values of the alpha 3-helix in the protein. The structural autonomy of the alpha 3-helix is profoundly determined by the presence of the conserved capping box and, in part, by the ionic bond formed between Glu200 and Lys204. On the basis of these observations a novel PrP consensus pattern, centered on the alpha 3-helix region, has been defined. The data indicate that this autonomous and highly conserved region of the PrPc likely plays a critical role in folding and stability. This gives an explanation of why many of pathogenic mutations occur in this part of the molecule, sharing relevant effects on the overall protein conformation. In particular the D202N capping mutation almost completely destabilizes the isolated alpha 3 peptide. While it is well known that the D202N substitution is associated with a GSS disease, the possible structural basis of this fatal pathology has never been investigated. We propose that a lower alpha 3-helical propensity leading to a major destabilization of the PrPc molecule initiates the pathogenic process associated with D202N capping mutation.


Asunto(s)
Mutación/fisiología , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Priones/genética , Priones/metabolismo , Secuencia de Aminoácidos , Animales , Dicroismo Circular , Humanos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Conformación Molecular , Datos de Secuencia Molecular , Fragmentos de Péptidos/síntesis química , Priones/síntesis química , Conformación Proteica
12.
Mol Endocrinol ; 13(1): 24-37, 1999 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9892010

RESUMEN

Hormones and growth factors regulate cell growth via the mitogen-activated protein (MAP) kinase cascade. Here we examine the actions of the hormone somatostatin on the MAP kinase cascade through one of its two major receptor subtypes, the somatostatin receptor 1 (SSTR1) stably expressed in CHO-K1 cells. Somatostatin antagonizes the proliferative effects of fibroblast growth factor in CHO-SSTR1 cells via the SSTR1 receptor. However, in these cells, somatostatin robustly activates MAP kinase (also called extracellular signal regulated kinase; ERK) and augments fibroblast growth factor-stimulated ERK activity. We show that the activation of ERK via SSTR1 is pertussis toxin sensitive and requires the small G protein Ras, phosphatidylinositol 3-kinase, the serine/threonine kinase Raf-1, and the protein tyrosine phosphatase SHP-2. The activation of ERK by SSTR1 increased the expression of the cyclin-dependent protein kinase inhibitor p21(cip1/WAF1). Previous studies have suggested that somatostatin-stimulated protein tyrosine phosphatase activity mediates the growth effects of somatostatin. Our data suggest that SHP-2 stimulation by SSTR1 may mediate some of these effects through the activation of the MAP kinase cascade and the expression of p21(cip1/WAF1).


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Proteínas Quinasas Activadas por Mitógenos , Receptores de Somatostatina/metabolismo , Somatostatina/metabolismo , Animales , Células CHO/efectos de los fármacos , Células CHO/metabolismo , Proteínas Quinasas Dependientes de Calcio-Calmodulina/efectos de los fármacos , División Celular , Cricetinae , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/efectos de los fármacos , Ciclinas/metabolismo , Activación Enzimática/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Proteínas de Unión al GTP/efectos de los fármacos , Proteínas de Unión al GTP/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Proteína Quinasa 3 Activada por Mitógenos , Toxina del Pertussis , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas Proto-Oncogénicas c-raf/metabolismo , Receptores de Somatostatina/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Tirosina Fosfatasas con Dominio SH2 , Somatostatina/farmacología , Factores de Virulencia de Bordetella/farmacología , Proteínas ras/genética , Proteínas ras/metabolismo , Dominios Homologos src
13.
Mol Endocrinol ; 8(10): 1289-97, 1994 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-7854346

RESUMEN

Somatostatin receptors are abundantly expressed on a variety of human endocrine and epithelial tumors. The ability of these receptors to couple to effector pathways that inhibit the growth of these tumor cells has prompted the use of somatostatin agonists in the treatment of human neoplasms. It has been demonstrated that somatostatin stimulates a phosphotyrosine phosphatase in human tumor cells through a receptor-mediated process. This stimulation may counteract the growth-promoting properties of growth factors and the receptor tyrosine kinases that they activate. The recent cloning and characterization of distinct somatostatin receptor subtypes raise the possibility that different receptor subtypes mediate distinct effector pathways. To determine whether cloned somatostatin receptors could mediate coupling to phosphotyrosine phosphotyrosine phosphatase activity, we examined phosphatase activity after somatotostatin activation of the rat somatostatin receptors SSTR1 and SSTR2 after their stable expression in heterologous Chinese Hamster Ovary (CHO-K1) cells. We found that stimulation of SSTR1 cells was capable of increasing phosphotyrosine phosphatase activity, despite the coupling of both receptors to the inhibition of adenylyl cyclase in these cells. This activation was characterized by an EC50 of 70 nM and was sensitive to pertussis toxin. In addition, we demonstrate that activation of phosphotyrosine phosphatase activity in pituitary cell lines correlates with the endogenous expression of the SSTR1 gene within these cells.


Asunto(s)
Proteínas Tirosina Fosfatasas/metabolismo , Receptores de Somatostatina/metabolismo , Toxina de Adenilato Ciclasa , Animales , Células CHO , Clonación Molecular , Cricetinae , ADN Complementario/genética , Activación Enzimática , Técnicas de Transferencia de Gen , Toxina del Pertussis , Ratas , Proteínas Recombinantes/metabolismo , Somatostatina/farmacología , Factores de Virulencia de Bordetella/farmacología
14.
Mol Endocrinol ; 15(10): 1838-52, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11579215

RESUMEN

The aim of this study was the characterization of the intracellular effectors of the antiproliferative activity of somatostatin in PC Cl3 thyroid cells. Somatostatin inhibited PC Cl3 cell proliferation through the activation of a membrane phosphotyrosine phosphatase. Conversely, PC Cl3 cells stably expressing the v-mos oncogene (PC mos) were completely insensitive to the somatostatin antiproliferative effects since somatostatin was unable to stimulate a phosphotyrosine phosphatase activity. In PC mos cells basal phosphotyrosine phosphatase activity was also reduced, suggesting that the expression of a specific phosphotyrosine phosphatase was impaired in these transformed cells. We suggested that this phosphotyrosine phosphatase could be r-PTP eta whose expression was abolished in the PC mos cells. To directly prove the involvement of r-PTP eta in somatostatin's effect, we stably transfected this phosphatase in PC mos cells. This new cell line (PC mos/PTP eta) recovered somatostatin's ability to inhibit cell proliferation, showing dose-dependence and time course similar to those observed in PC Cl3 cells. Conversely, the transfection of a catalytically inactive mutant of r-PTP eta did not restore the antiproliferative effects of somatostatin. PC mos/PTP eta cells showed a high basal phosphotyrosine phosphatase activity which, similarly to PC Cl3 cells, was further increased after somatostatin treatment. The specificity of the role of r-PTP eta in somatostatin receptor signal transduction was demonstrated by measuring its specific activity after somatostatin treatment in an immunocomplex assay. Somatostatin highly increased r-PTP eta activity in PCCl3 and PC mos/PTP eta (+300%, P < 0.01) but not in PCmos cells. Conversely, no differences in somatostatin-stimulated SHP-2 activity, (approximately +50%, P < 0.05), were observed among all the cell lines. The activation of r-PTP eta by somatostatin caused, acting downstream of MAPK kinase, an inhibition of insulin-induced ERK1/2 activation with the subsequent blockade of the phosphorylation, ubiquitination, and proteasome degradation of the cyclin-dependent kinase inhibitor p27(kip1). Ultimately, high levels of p27(kip1) lead to cell proliferation arrest. In conclusion, somatostatin inhibition of PC Cl3 cell proliferation requires the activation of r-PTP eta which, through the inhibition of MAPK activity, causes the stabilization of the cell cycle inhibitor p27(kip1).


Asunto(s)
División Celular/efectos de los fármacos , Proteínas Tirosina Fosfatasas/metabolismo , Somatostatina/farmacología , Glándula Tiroides/citología , Northern Blotting , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Técnica del Anticuerpo Fluorescente , Proteínas de Unión al GTP/fisiología , Expresión Génica , Insulina/farmacología , Cinética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mutación , Fosforilación , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas/genética , ARN Mensajero/análisis , Receptores de Somatostatina/fisiología , Transducción de Señal , Tirotropina/farmacología , Transfección , Proteínas Supresoras de Tumor/metabolismo , Vanadatos/farmacología
15.
Endocrinology ; 137(10): 4409-18, 1996 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8828502

RESUMEN

cAMP and cyclosporin A exert antiproliferative effects in many different cell types. In cultured pituitary cells, the antiproliferative effects of both agents correlate with the inhibition of a serine/threonine protein phosphatase activity. This inhibition is mediated by the heat-stable protein, inhibitor-1. The increase of cAMP levels, through the activation of the protein kinase A, induces inhibitor-1 phosphorylation and activation. On the other hand, cyclosporin A, inhibiting the calcium-dependent serine/threonine phosphatase calcineurin, prevents the dephosphorylation and inactivation of inhibitor-1. This dual regulation by cAMP and calcium on the inhibitor-1 activity parallel the effects of these agents on DNA synthesis and serine/threonine phosphatase activity. Because inhibitor-1 is the main regulator of protein phosphatase 1 activity, these results suggest that protein phosphatase 1 may be the common target of cAMP and cyclosporin A in regulating cell proliferation. We propose that protein-phosphatase 1 stimulates growth in these cells and that cAMP and cyclosporin A block this effect through their actions on inhibitor-1.


Asunto(s)
AMP Cíclico/farmacología , Ciclosporina/farmacología , Fosfoproteínas Fosfatasas/metabolismo , Hipófisis/metabolismo , División Celular/efectos de los fármacos , Línea Celular , Dopamina/farmacología , Inhibidores Enzimáticos/metabolismo , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/fisiología , Hipófisis/citología , Hipófisis/efectos de los fármacos , Proteína Fosfatasa 1 , Proteínas Tirosina Fosfatasas/metabolismo
16.
Endocrinology ; 126(3): 1435-41, 1990 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-2407518

RESUMEN

Recent findings indicate that interleukin-1 beta (IL1 beta), a monokine secreted by stimulated macrophages and monocytes, modulates neuroendocrine functions in a manner similar to classical hormones. In this study we show that IL1 modulates PRL secretion, assessed by reverse hemolytic plaque assay, and describe the effect of the monokine on adenylate cyclase activity and calcium fluxes in rat normal pituitary cells. In basal and vasoactive intestinal peptide (VIP)-stimulated conditions, low doses of IL1 reduced the mean plaque area, a direct index of PRL secretion without affecting the percentage of PRL-secreting cells. Similarly, low concentrations of IL1 inhibited adenylate cyclase activity in both basal and VIP-stimulated conditions, while higher concentrations restored the enzymatic activity to the control value. IL1 also caused a biphasic effect on the free intracellular calcium increase induced by maitotoxin, a calcium channel activator, being inhibitory at low and stimulatory at high concentrations. The effects of IL1 on adenylate cyclase activity and calcium fluxes were reversed by preincubation of the monokine with its polyclonal antibody, thus confirming the specificity of the effects. In conclusion, our data show that IL1 modulates PRL secretion by acting directly on pituitary cells through interaction with the adenylate cyclase-cAMP system and calcium flux.


Asunto(s)
Adenilil Ciclasas/metabolismo , Calcio/metabolismo , Interleucina-1/fisiología , Oxocinas , Adenohipófisis/metabolismo , Prolactina/metabolismo , Animales , Técnica de Placa Hemolítica , Toxinas Marinas/farmacología , Adenohipófisis/citología , Ratas , Péptido Intestinal Vasoactivo/farmacología
17.
Endocrinology ; 138(9): 3756-63, 1997 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9275062

RESUMEN

The effects of the stable expression of E1A and/or middle T oncogenes on the proliferative activity of PC Cl3 normal thyroid cells are reported. The proliferation of PC Cl3 cells is mainly regulated by insulin and TSH in a stimulatory way and by somatostatin in an inhibitory fashion. The transformed cell lines, named PC Py and PC E1A Py, show an autonomous pattern of proliferation. The blockade of phosphotyrosine phosphatase activity with vanadate increased the proliferation rate of PC Cl3 under basal and stimulated conditions and completely prevented the inhibitory activity of somatostatin, suggesting that in PC Cl3 cells, a tonic tyrosine phosphatase activity regulates basal and stimulated proliferation, and that a somatostatin-dependent increase in this activity may represent a cytostatic signal. Conversely, in both PC Py and PC E1A Py, vanadate did not modify basal and stimulated proliferation. We analyzed tyrosine phosphatase activity in the different cell lines basally and under conditions leading to the arrest of cell proliferation: confluence (contact inhibition), growth factor deprivation (starvation), and somatostatin treatment. Under basal conditions, tyrosine phosphatase activity was significantly lower in PC Py and PC E1APy cell lines than that in the normal cells. The inhibition of the proliferation induced by contact inhibition or somatostatin treatment was accompanied by an increase in tyrosine phosphatase activity only in PC Cl3 cells. The reduction in tyrosine phosphatase activity in PC E1APy cells correlated with a significant reduction in the expression of R-PTP eta, a tyrosine phosphatase cloned from PC Cl3 cells. Conversely, the expression of another receptor-like PTP, PTP mu, was unchanged. Thus, PTP eta may be a candidate to mediate inhibitory signals (i.e. activation of somatostatin receptors or cell to cell contact) on the proliferative activity of PC Cl3 cells, and the reduction of its expression in the transformed cell lines may lead to an alteration in the control of cell proliferation.


Asunto(s)
División Celular , Transformación Celular Neoplásica/genética , Oncogenes , Proteínas Tirosina Fosfatasas/metabolismo , Glándula Tiroides/citología , Glándula Tiroides/enzimología , Animales , Northern Blotting , Línea Celular , Células Clonales , Medios de Cultivo , ADN/biosíntesis , Inhibidores Enzimáticos/farmacología , Citometría de Flujo , Expresión Génica , Sustancias de Crecimiento/administración & dosificación , Humanos , Insulina/farmacología , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Proteínas Tirosina Fosfatasas/genética , Ratas , Somatostatina/farmacología , Vanadatos/farmacología
18.
Endocrinology ; 131(4): 1942-7, 1992 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-1356754

RESUMEN

The sulfonylurea glibenclamide, which is known to block ATP-sensitive potassium channels, increases, in a dose-dependent manner, the release of PRL from MMQ pituitary cells. Glibenclamide does not reduce the dopaminergic inhibition of forskolin-stimulated PRL secretion; conversely it almost completely abolishes the inhibitory effect of somatostatin (SRIF) on this parameter. The sulfonylurea dose dependently increases basal [Ca++]i, without affecting the increase in [Ca++]i induced by high concentrations of extracellular potassium. Glibenclamide does not modify dopamine-induced [Ca++]i reduction, whereas it abolishes the inhibitory effect of SRIF on basal [Ca++]i. In the presence of diazoxide, an opener of ATP-sensitive potassium channels, which lowers basal [Ca++]i, dopamine still reduces [Ca++]i whereas SRIF does not induce a further decrease. Glibenclamide induces the depolarization of the cell membrane and prevents the SRIF-evoked hyperpolarization. The hyperpolarization of the cell membrane induced by dopamine is not modified by glibenclamide. Diazoxide induces a cell membrane hyperpolarization that is enhanced by dopamine but not by SRIF. Finally, glibenclamide does not affect basal and stimulated adenylate cyclase activity. In conclusion, our findings show that, in MMQ cells, glibenclamide stimulates PRL release, suggesting an involvement of ATP-sensitive potassium channels in the regulation of PRL secretion. The reversal by glibenclamide of the effects of SRIF on calcium homeostasis, membrane potential, and PRL release suggests that this type of potassium channel participates to the somatostatinergic inhibition of PRL secretion. Conversely, we found that glibenclamide does not modify the dopaminergic inhibition of PRL secretion and second messenger systems, suggesting that ATP-sensitive potassium channels may not be involved in the inhibitory effect of dopamine on PRL release.


Asunto(s)
Adenosina Trifosfato/farmacología , Dopamina/farmacología , Hipófisis/metabolismo , Canales de Potasio/fisiología , Prolactina/antagonistas & inhibidores , Somatostatina/farmacología , Calcio/metabolismo , Línea Celular , Diazóxido/farmacología , Gliburida/farmacología , Hipófisis/citología , Potasio/farmacología , Canales de Potasio/efectos de los fármacos , Prolactina/metabolismo
19.
Endocrinology ; 144(4): 1574-84, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12639942

RESUMEN

Somatostatin was reported to inhibit Kaposi's sarcoma (KS) cell (KS-Imm) xenografts through an antiangiogenic activity. Here, we show that somatostatin blocks growth of established KS-Imm tumors with the same efficacy as adriamycin, a clinically effective cytotoxic drug. Whereas KS-Imm cells do not express somatostatin receptors (SSTRs), endothelial cells express several SSTRs, in particular SSTR3. We investigated the molecular mechanisms and receptor specificity of somatostatin inhibition of angiogenesis. Somatostatin significantly inhibited angiogenesis in vivo in the matrigel sponge assay; this inhibition was mimicked by the SSTR3 agonist L-796778 and reversed by the SSTR3 antagonist BN81658, demonstrating involvement of SSTR3. In vitro experiments showed that somatostatin directly affected different endothelial cell line proliferation through a block of growth-factor-stimulated MAPK and endothelial nitric oxide (NO) synthase (eNOS) activities. BN81658 reversed somatostatin inhibition of cell proliferation, NO production, and MAPK activity, indicating that SSTR3 activation is required for the effects of somatostatin in vitro. Finally in vivo angiogenesis assays demonstrated that eNOS inhibition was a prerequisite for the antiangiogenic effects of somatostatin, because high concentrations of sodium nitroprusside, an NO donor, abolished the somatostatin effects. In conclusion, we demonstrate that somatostatin is a powerful antitumor agent in vivo that inhibits tumor angiogenesis through SSTR3-mediated inhibition of both eNOS and MAPK activities.


Asunto(s)
Hormonas/farmacología , Neovascularización Patológica/tratamiento farmacológico , Óxido Nítrico Sintasa/metabolismo , Receptores de Somatostatina/metabolismo , Somatostatina/farmacología , Amidas/farmacología , Animales , División Celular/efectos de los fármacos , Células Cultivadas , Endotelio Vascular/citología , Endotelio Vascular/enzimología , Activación Enzimática/efectos de los fármacos , Humanos , Técnicas In Vitro , Ratones , Ratones Desnudos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico Sintasa de Tipo III , Nitrobencenos/farmacología , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/antagonistas & inhibidores , Venas Umbilicales/citología
20.
Endocrinology ; 144(10): 4298-305, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12959980

RESUMEN

The Src homology 2-containing tyrosine phosphatase, Shp-2, is a crucial enzyme that mediates intracellular signaling and is implicated in cell proliferation and differentiation. Here we investigated the involvement of the Shp-2 tyrosine phosphatase in determining the downstream signaling pathways initiated by the Ret oncogene, carrying either the cysteine 634 to tyrosine or the methionine 918 to threonine substitutions. These mutations convert the receptor tyrosine kinase, Ret, into a dominant transforming protein and induce constitutive activation of its intrinsic tyrosine kinase activity leading to congenital and sporadic cancers in neuroendocrine organs. Using the PC12, rat pheochromocytoma cell line, as model system, we show that Shp-2 mediates immediate-early gene expression if induced by either of the mutant alleles. Furthermore, we show that Shp-2 activity is required for RetM918T-induced Akt activation. The results indicate that Shp-2 is a downstream mediator of the mutated receptors RetC634Y and RetM918T, thus suggesting that it may act as a limiting factor in Ret-associated endocrine tumors, in the neoplastic syndromes multiple endocrine neoplasia types 2A and 2B.


Asunto(s)
Membranas Intracelulares/fisiología , Mutación/fisiología , Proteínas Oncogénicas/genética , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Transducción de Señal/fisiología , Animales , Diferenciación Celular/fisiología , Línea Celular , Supervivencia Celular/fisiología , Factor Neurotrófico Derivado de la Línea Celular Glial , Péptidos y Proteínas de Señalización Intracelular , Factores de Crecimiento Nervioso/metabolismo , Proteínas Oncogénicas/metabolismo , Células PC12/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteínas Proto-Oncogénicas c-ret , Ratas , Proteínas Tirosina Quinasas Receptoras/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA