Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 245
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Neuroscience ; 158(4): 1731-41, 2009 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-19041374

RESUMEN

Epidermal growth factor (EGF) has a neurotrophic activity on developing midbrain dopaminergic neurons. We investigated developmental effects of peripheral EGF administration on dopaminergic neurons in midbrain slice preparations containing ventral tegmental area (VTA). Subcutaneous EGF administration to mouse neonates triggered phosphorylation of EGF receptors (ErbB1 and ErbB2) in the midbrain region, suggesting its penetration through the blood-brain barrier. We repeated EGF administration in postnatal mice and examined synaptic transmission in the VTA with electrophysiological recordings. Subchronic EGF treatment increased the amplitude of field excitatory postsynaptic potentials evoked by stimulation of the anterior VTA. To analyze the EGF effect at a single cell level, dopaminergic neurons were identified by their characteristic hyperpolarizing activated currents in whole cell recording. In these dopaminergic neurons, EGF effects the amplitude of spontaneous miniature excitatory postsynaptic currents (mEPSCs) without affecting their frequency. In agreement, EGF also enhanced the AMPA/NMDA ratio of evoked EPSCs in the dopaminergic neurons. In contrast, EGF effects on mEPSCs of neighboring neurons not exhibiting hyperpolarizing activated currents were modest or insignificant. Thus, these results suggest that circulating EGF substantially influences the physiological properties of developing midbrain dopaminergic neurons in perinatal and postnatal mice.


Asunto(s)
Dopamina/metabolismo , Factor de Crecimiento Epidérmico/administración & dosificación , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Área Tegmental Ventral/citología , Animales , Animales Recién Nacidos , Biofisica , Biotinilación/métodos , Estimulación Eléctrica/métodos , Receptores ErbB/genética , Receptores ErbB/metabolismo , Potenciales Postsinápticos Excitadores/fisiología , Femenino , Humanos , Técnicas In Vitro , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos C57BL , Técnicas de Placa-Clamp/métodos , Fosforilación , Embarazo , Receptores AMPA/genética , Receptores AMPA/metabolismo , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Área Tegmental Ventral/efectos de los fármacos , Área Tegmental Ventral/metabolismo
3.
J Clin Invest ; 86(1): 120-5, 1990 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-1973172

RESUMEN

We determined clonality of thyroid tumors from female patients who had restriction fragment length polymorphisms (RFLP) in the X chromosome genes hypoxanthine phosphoribosyltransferase (HPRT) or phosphoglycerate kinase (PGK). We screened normal thyroid tissue from 59 female patients; of the informative cases 14 were heterozygous for a Bgl I site on PGK and 4 were heterozygous for a Bam HI site on HPRT. In monoclonal tumors, one of the polymorphic alleles was selectively digested after additional digestion with Hpa II, a methylation sensitive enzyme, whereas in polyclonal tissue both were decreased to a similar extent. Normal thyroid tissue from all patients showed a polyclonal pattern. Of the 18 tumors studied, 12 were solitary thyroid nodules, and 6 were obtained from multinodular goiters (MNG). The following were monoclonal: 6/6 follicular adenomas, 2/2 follicular carcinomas, and 1/1 anaplastic carcinoma. Two of the three papillary carcinomas showed intermediate patterns, possibly due to contaminating effects of stromal tissue present in most of these neoplasms. Of the six nodules from MNG, four were polyclonal. The two largest gave a distinct monoclonal pattern. Most solitary thyroid tumors are monoclonal, supporting a somatic cell mutation model of thyroid neoplasm formation. Nodules from MNG are largely hyperplastic, although monoclonal neoplasms do occasionally arise within these glands. The specific somatic mutations leading to clonal expansion and determination of tumor phenotype are presently unknown.


Asunto(s)
Neoplasias de la Tiroides/patología , Adenoma/patología , Carcinoma/patología , Células Clonales , ADN de Neoplasias/genética , Compensación de Dosificación (Genética) , Femenino , Bocio/patología , Humanos , Hipoxantina Fosforribosiltransferasa/genética , Fosfoglicerato Quinasa/genética , Polimorfismo de Longitud del Fragmento de Restricción , Cromosoma X/fisiología
4.
Radiat Res ; 165(1): 35-42, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16392960

RESUMEN

Gleevec, a selective tyrosine kinase inhibitor, retarded the growth of anaplastic thyroid cancer cell lines in vitro and in vivo through selective inhibition of ABL tyrosine kinase activity. In the present study, we investigated the ability of Gleevec to modulate the in vitro and in vivo radiation response of anaplastic thyroid cancer cells. Cell growth assays, colony formation assays and xenograft models were used to quantify the radiosensitizing effect of Gleevec in cells of the anaplastic thyroid cancer cell lines ARO and FRO. FACS, Western blotting and histochemical techniques were employed to study the mechanisms of radiation response after exposure to Gleevec. Gleevec (7.0 microM) increased the anti-proliferative effect of radiation on the growth ARO and FRO cells in vitro. Clonogenic analysis demonstrated that Gleevec reduced cell survival after irradiation. Gleevec combined with radiation produced an increase in tumor growth inhibition compared to treatment with either modality alone in mice bearing anaplastic thyroid cancer xenografts. The drug suppressed radiation-induced ABL activation and promoted CDKN1A (p21(cip1)) accumulation in irradiated anaplastic thyroid cancer cells. Gleevec had an additional effect on radiation-induced apoptosis in cells of both cell lines and potentiated the induction of terminal growth arrest accompanied by the expression of senescence-associated beta-galactosidase. The antitumor effect of Gleevec is potentiated in adjunctive therapy with radiation not only due to inhibition of proliferative cell growth with transient cell cycle arrest and apoptosis, but also due to the terminal growth arrest associated with senescence, suggesting that tumor cell senescence is a mechanism for tumor targeting therapy in combination with ionizing radiation.


Asunto(s)
Carcinoma/enzimología , Carcinoma/patología , Piperazinas/administración & dosificación , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Pirimidinas/administración & dosificación , Neoplasias de la Tiroides/enzimología , Neoplasias de la Tiroides/patología , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Benzamidas , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Proteínas de Fusión bcr-abl , Humanos , Mesilato de Imatinib , Dosis de Radiación , Tolerancia a Radiación/efectos de los fármacos
6.
Cancer Res ; 59(13): 3049-52, 1999 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-10397242

RESUMEN

Our recent study demonstrates the feasibility of the thyroglobulin (TG) promoter in transcriptionally targeted gene therapy for thyroid carcinomas expressing TG, albeit less effectively than the constitutive viral promoter. The present study was, therefore, designed to enhance the activity of the TG promoter with the Cre-loxP system. Our data demonstrate that the in vitro cytotoxic effect of herpes simplex virus thymidine kinase/ganciclovir obtained with the TG promoter and the Cre-loxP system is approximately 5-10-fold higher than that with the TG promoter alone. Enhanced tumor growth inhibition was also observed in in vivo tumor models. These data indicate the usefulness of the Cre-loxP system to enhance the activity of a tissue (or tumor)-specific promoter in transcriptionally targeted cancer gene therapy.


Asunto(s)
Marcación de Gen , Terapia Genética/métodos , Tiroglobulina/genética , Neoplasias de la Tiroides/terapia , Animales , Línea Celular , Línea Celular Transformada , Ganciclovir/uso terapéutico , Genes Reporteros , Vectores Genéticos , Humanos , Ratones , Ratones Desnudos , Profármacos/uso terapéutico , Regiones Promotoras Genéticas , Ratas , Proteínas Recombinantes/biosíntesis , Retroviridae , Simplexvirus/genética , Timidina Quinasa/genética , Glándula Tiroides , Transcripción Genética , Transfección
7.
Cancer Res ; 55(10): 2075-80, 1995 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-7743505

RESUMEN

We investigated the sensitivity and sequential events that take place in thyroid epithelial cells after irradiation. Cell survival ratios at a dose of 2 Gy were 18 +/- 2.5%, 58 +/- 1.0%, 59 +/- 1.5%, and 98 +/- 1.8% in primary thyroid cells, papillary thyroid carcinoma cells, follicular thyroid carcinoma cells, and anaplastic thyroid carcinoma cells, respectively. Thyroid carcinoma cell lines carrying mutations in the p53 gene were resistant to ionizing radiation. Although irradiated cells were accumulated at G1 in primary thyroid cells even after low-dose irradiation (0.2 Gy), this phenomenon was not observed in the thyroid carcinoma cell lines. Wild-type p53 expression in primary thyroid cell was increased following irradiation, but mutated p53 in the thyroid carcinoma cell lines was unchanged. To clarify the signal transduction in the G1 arrest following irradiation, levels of expression of the p53 putative downstream effectors GADD45 and WAF1/Cip1 were examined. Despite the consistent level of GADD45 mRNA, the level of WAF1/Cip1 transcripts was increased in a radiation dose-dependent manner in primary thyroid cells. This increase in the WAF1/Cip1 mRNA level was observed 30 min after irradiation and continued for at least 48 h. A mobility shift assay performed using the sequence of the putative p53 DNA binding site on the WAF1/Cip1 and GADD45 genes as a probe showed that nuclear protein extracted from primary thyroid cells, anti-p53 antibody, and probe oligonucleotide-bound complex was clearly shifted. An increase in binding activity of the p53/antibody/DNA complex was observed following irradiation. In contrast, the nuclear extract from thyroid carcinoma cells could not bind the specific DNA site, suggesting that mutant p53 has lost its binding ability. Actinomycin D inhibited WAF1/Cip1 and GADD45 mRNA levels and cycloheximide stimulated up-regulation of both basal mRNA levels, but an additional increase of the mRNA expression following irradiation was observed only in the WAF1/Cip1 gene. These data suggest that p53 in postradiation acts at a transcriptional level on WAF1/Cip1 gene expression and that de novo protein synthesis is not required for this effect. These results suggest that the p53-WAF1/Cip1 pathway may play a central role in induction of G1 arrest following irradiation in human thyroid epithelial cells.


Asunto(s)
Carcinoma Papilar/metabolismo , Carcinoma/metabolismo , Fase G1 , Genes p53/efectos de la radiación , Glándula Tiroides/metabolismo , Neoplasias de la Tiroides/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Secuencia de Bases , Línea Celular , Cicloheximida/farmacología , Dactinomicina/farmacología , Fase G1/efectos de la radiación , Genes p53/fisiología , Humanos , Datos de Secuencia Molecular , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Glándula Tiroides/citología , Glándula Tiroides/efectos de la radiación
8.
Cancer Res ; 56(1): 77-86, 1996 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-8548779

RESUMEN

A newly established metastatic rat pituitary tumor (mGH3) possesses a malignant phenotype that is invasive and hypervascular compared with the original GH3 tumors. mGH3 cells exhibit anchorage independence and expression of elevated levels of parathyroid hormone-related peptide (PTHrP) in vitro. To clarify the role of PTHrP in the development of the malignant phenotype, tumor cells were treated with phosphorothioate antisense PTHrP oligonucleotide. Treatment with antisense PTHrP resulted in a scattering phenomenon in the colony formation assay but did not inhibit cell growth in vitro. Inoculation of mGH3 cells in the cerebral ventricle resulted in a rapid growth of tumor cells within 3 weeks and dissemination throughout the entire ventricular system. Although treatment with sense or mismatched PTHrP oligonucleotide did not influence the subsequent tumor growth, the in vivo coinjection and injection of antisense PTHrP 1 week after tumor cell implantation into the right lateral ventricle markedly reduced tumor size and suppressed metastasis formation. The survival rate of mGH3 tumor-injected rats was prolonged by antisense PTHrP therapy. Our results demonstrated the biological involvement of PTHrP in malignant phenotype in rat pituitary tumors, suggesting that antisense PTHrP may provide a novel antimetastatic therapy for malignant somatotroph tumors.


Asunto(s)
Expresión Génica/efectos de los fármacos , Oligonucleótidos Antisentido/farmacología , Neoplasias Hipofisarias/genética , Proteínas/antagonistas & inhibidores , Animales , Secuencia de Bases , División Celular/efectos de los fármacos , Femenino , Datos de Secuencia Molecular , Metástasis de la Neoplasia , Hormona Paratiroidea/metabolismo , Proteína Relacionada con la Hormona Paratiroidea , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología , Proteínas/genética , Ratas , Células Tumorales Cultivadas
9.
AJNR Am J Neuroradiol ; 37(10): 1851-1859, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27339950

RESUMEN

BACKGROUND AND PURPOSE: The stiffness of intracranial tumors affects the outcome of tumor removal. We evaluated the stiffness of 4 common intracranial tumors by using MR elastography and tested whether MR elastography had the potential to discriminate firm tumors preoperatively. MATERIALS AND METHODS: Thirty-four patients with meningiomas, pituitary adenomas, vestibular schwannomas, and gliomas scheduled for resection were recruited for MR elastography. On the elastogram, the mean and the maximum shear stiffnesses were measured by placing an ROI on the tumor. Blinded to the MR elastography findings, surgeons conducted qualitative intraoperative assessment of tumor consistency by using a 5-point scale. Histopathologic diagnosis was confirmed by using the resected specimens. The mean and maximum shear stiffnesses were compared with histopathologic subtypes, and the intraoperative tumor consistency was graded by the surgeons. RESULTS: The mean and maximum shear stiffnesses were the following: 1.9 ± 0.8 kPa and 3.4 ± 1.5 kPa for meningiomas, 1.2 ± 0.3 kPa and 1.8 ± 0.5 kPa for pituitary adenomas, 2.0 ± 0.4 kPa and 2.7 ± 0.8 kPa for vestibular schwannomas, and 1.5 ± 0.2 kPa and 2.7 ± 0.8 kPa for gliomas. The mean and maximum shear stiffnesses for meningiomas were higher than those of pituitary adenomas (P < .05). The mean and maximum shear stiffnesses were significantly correlated with the surgeon's qualitative assessment of tumor consistency (P < .05). The maximum shear stiffness for 5 firm tumors was higher than that of nonfirm tumors (P < .05). CONCLUSIONS: MR elastography could evaluate intracranial tumors on the basis of their physical property of shear stiffness. MR elastography may be useful in discriminating firm tumors preoperatively.

10.
Oncogene ; 20(8): 989-96, 2001 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-11314034

RESUMEN

The thyroid gland is one of the most sensitive organs in ionizing radiation (IR)-induced carcinogenesis. To determine, therefore, the specific cascade of IR-induced signal transduction in human thyroid cells, we investigated the functional role of protein kinase C (PKC), especially its interlocking activation of c-Jun NH(2)-terminal kinase (JNK) pathway. In the present study, using adenovirus expression vectors for diverse dominant-negative (DN) types of PKC isoforms (alpha, beta2, delta, epsilon and zeta) expressed in primary cultured human thyroid cells, only DN/PKC delta suppressed IR-induced JNK activation. In addition, Rottlerin, a PKC delta specific inhibitor, inhibited IR-induced JNK activation. IR-induced activation of transcription factor AP-1, downstream target of JNK, was also attenuated by DN/PKC delta. To examine the involvement of upstream kinases of JNK, we performed immune-complex kinase assays of mitogen-activated protein kinase kinase 4 (MKK4) and MKK7. IR activated MKK7 but not MKK4, and this activation was inhibited by Rottlerin. Furthermore, IR-induced JNK activation was suppressed by overexpression of kinase-deficient MKK7. Our results indicate that IR selectively activates the cascade of PKC delta-MKK7-JNK-AP-1 in human thyroid cells, suggesting a not apoptotic but radio-resistant role of PKC delta in human thyroid cells following IR.


Asunto(s)
Isoenzimas/metabolismo , MAP Quinasa Quinasa 4 , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteína Quinasa C/metabolismo , Glándula Tiroides/efectos de la radiación , Activación Enzimática , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos , MAP Quinasa Quinasa 7 , Fosforilación , Proteína Quinasa C-delta , Radiación Ionizante , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción AP-1/metabolismo , Tirosina/metabolismo , Rayos Ultravioleta , Rayos X
11.
Oncogene ; 14(13): 1511-9, 1997 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-9129141

RESUMEN

To understand the effects of ionizing radiation on thyroid cells, we investigated the role of p53 in mediating apoptosis and in DNA repair following in vivo and in vitro irradiation of thyroid cells. In vitro exposure of human thyroid cells to ionizing radiation of up to 5-8 Gy failed to induce apoptosis in primary cells. The same results were obtained when the thyroid gland was irradiated in the intact rat. To explore the mechanism of failure of the wild-type p53 in inducing apoptosis in thyroid cells, we investigated the expression of apoptosis-related genes, bax, bcl-2 and fas/APO-1 following irradiation or induction of temperature-sensitive p53. The expression of Bax, Bcl-2 and Fas/APO-1 in human primary cultured thyroid cells did not change after irradiation. To further confirm the results, we established a clonal cell line (tsFRO) in which a temperature sensitive p53 (Val138) expression vector was stably transfected to a thyroid carcinoma cell line lacking endogenous p53. Incubation of tsFRO cells at the permissive temperature for three days, however, did not induce apoptosis although G1 arrest was noted. Although enhanced expression of the bax mRNA level was observed, the expression of Bax, Bcl-2 and Fas/APO-1 protein did not change by shifting tsFRO cells to permissive temperature as well as irradiated primary cells. Furthermore, DNA end-jointing ability was examined by transfection of linearized luciferase plasmid into tsFRO cells. Increased luciferase activity occurred when the cells were cultured at the permissive temperature, indicating that the wild-type p53 enhances DNA end-jointing activity. Our results indicate that the wild-type p53 does not lead to apoptosis but facilitates DNA end-jointing in thyroid cells. These results may reflect specific responses in thyroid cells following irradiation.


Asunto(s)
Apoptosis , Reparación del ADN , ADN/metabolismo , Genes p53 , Proteínas Proto-Oncogénicas c-bcl-2 , Radiación Ionizante , Glándula Tiroides/citología , Glándula Tiroides/metabolismo , Animales , Células Cultivadas , Fase G1 , Regulación de la Expresión Génica , Humanos , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas/genética , Ratas , Ratas Endogámicas WF , Temperatura , Transfección , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/fisiología , Proteína X Asociada a bcl-2
12.
Mol Endocrinol ; 4(10): 1474-9, 1990 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-2283998

RESUMEN

Identifying the nature of the genetic mutations in thyroid neoplasms and their prevalence in the various tumor phenotypes is critical to understanding their pathogenesis. Mutational activation of ras oncogenes in human tumors occurs predominantly through point mutations in two functional regions of the molecules, codons 12, 13 (GTP-binding domain) or codon 61 (GTPase domain). We examined the prevalence of point mutations in codons 12, 13, and 61 of the oncogenes K-ras, N-ras, and H-ras in benign and malignant human thyroid tumors by hybridization of PCR-amplified tumor DNA with synthetic oligodeoxynucleotide probes. None of the eight normal thyroid tissues harbored point mutations. Four of nineteen nodules from multinodular goiters (21%), 6/24 microfollicular adenomas (25%), 3/14 papillary carcinomas (21%), and 0/3 follicular carcinomas contained ras point mutations. The predominant mutation was a valine for glycine substitution in codon 12 of H-ras. None of the multinodular goiter tumors known to be polyclonal (and thus due to hyperplasia) had point mutations, whereas one of the two monoclonal adenomas arising in nodular glands contained in H-ras codon 12 valine substitution, which was confirmed by sequencing the tumor DNA. These data show that ras activation is about equally prevalent in benign and malignant thyroid neoplasms, and thus may be an early event in the tumorigenic process.


Asunto(s)
Genes ras/genética , Mutación , Neoplasias de la Tiroides/genética , Adenocarcinoma/genética , Adenoma/genética , Secuencia de Bases , Carcinoma Papilar/genética , Codón , ADN de Neoplasias/genética , Bocio Nodular/genética , Humanos , Datos de Secuencia Molecular , Hibridación de Ácido Nucleico , Sondas de Oligonucleótidos , Reacción en Cadena de la Polimerasa
13.
Curr Mol Med ; 15(3): 222-36, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25817857

RESUMEN

Neuregulin-1 (NRG1) is a well-recognized risk gene for schizophrenia and is often implicated in the neurodevelopmental hypothesis of this illness. Alternative splicing and proteolytic processing of the NRG1 gene produce more than 30 structural variants; however, the neuropathological roles of individual variants remain to be characterized. On the basis of the neurodevelopmental hypothesis of schizophrenia, we administered eNRG1 (0.1~1.0 µg/g), a core epidermal growth factor-like (EGF) domain common for all splicing NRG1 variants, to neonatal mice and compared their behavioral performance with mice challenged with a full mature form of type 1 NRG1 variant. During the neonatal stage, recombinant eNRG1 protein administrated from the periphery passed the blood-brain barrier and activated its receptor (ErbB4) in the brain. In adults, the mice receiving the highest dose exhibited lower locomotor activity and deficits in prepulse inhibition and tonedependent fear learning, although the hearing reduction of the eNRG1-treated mice may explain these behavioral deficits. Neonatal eNRG1 treatment also significantly potentiated MK-801-driven locomotor activity in an eNRG1 dose-dependent manner. In parallel eNRG1 treatment enhanced MK-801-driven c-Fos induction and decreased immunoreactivity for NMDA receptor subunits in adult brain. In contrast, mice that had been treated with the same molar dose of a full mature form of type 1 NRG1 as neonates did not exhibit hypersensitivity to MK-801. However, both animal models exhibited similar hypersensitivity to methamphetamine. Collectively, our findings suggest that aberrant peripheral NRG1 signals during neurodevelopment alter later behavioral traits and auditory functions in the NRG1 subtype-dependent manner.


Asunto(s)
Maleato de Dizocilpina/farmacología , Neurregulina-1/uso terapéutico , Esquizofrenia/tratamiento farmacológico , Animales , Animales Recién Nacidos , Encéfalo/metabolismo , Maleato de Dizocilpina/uso terapéutico , Evaluación Preclínica de Medicamentos , Femenino , Masculino , Ratones Endogámicos C57BL , Actividad Motora/efectos de los fármacos , Neurregulina-1/farmacocinética , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Receptores de N-Metil-D-Aspartato/metabolismo , Distribución Tisular
14.
Hum Gene Ther ; 7(15): 1847-52, 1996 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-8894676

RESUMEN

Antitumor effects of herpes simplex virus-thymidine kinase (HSV-tk) gene transfer followed by ganciclovir (GCV) administration were studied by serial magnetic resonance imaging (MRI) with reference to the bystander effect. Mixed populations of 9L-gliosarcoma cells transduced with the HSV-tk gene (TK cells) and wild-type 9L cells were implanted into the brain of syngeneic Fisher rats at various ratios (total cell number, 10(5) cells; percentage of TK cells, 100%, 25%, 10%, or 0%). Rats were treated with GCV (30 mg/kg per day) or saline for 14 days and tumor masses were visually monitored using MRI. All of the saline-treated rats (regardless of TK cell percentage) and GCV-treated rats inoculated with 0% TK cells died between day 19 and day 31 (mean survival, 22.9 days) due to progressive tumor growth. The GCV-treated rats inoculated with more than 10% of TK cells lived significantly longer than the saline-treated rats (p < 0.01). The mean survivals of GCV-treated groups were 50.7, 70.0, and longer than 100 days for 10%, 25%, and 100% TK cells, respectively. MRI study revealed that reduction in tumor size and disappearance of tumor were observed in the GCV-treated rats inoculated with 10% or 25% TK cells. Complete regression of the tumor was, however, observed only in the rats implanted with 100% TK cells. The present results show that the bystander effect is clearly observed in vivo in a TK percentage-dependent manner, and a population of more than 25% of TK-positive cells is required for complete tumor elimination.


Asunto(s)
Neoplasias Encefálicas/patología , Simplexvirus , Timidina Quinasa/genética , Animales , Neoplasias Encefálicas/genética , Ganciclovir/uso terapéutico , Técnicas de Transferencia de Gen , Gliosarcoma/genética , Gliosarcoma/patología , Imagen por Resonancia Magnética , Reacción en Cadena de la Polimerasa , Ratas , Ratas Endogámicas F344
15.
Hum Gene Ther ; 9(1): 5-11, 1998 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-9458237

RESUMEN

Transfer of the herpes simplex virus-thymidine kinase (HSV-tk) gene, followed by administration of ganciclovir (GCV), generates the "bystander effect," in which HSV-tk-negative wild-type cells, as well as HSV-tk-expressing cells, are killed by GCV. To eradicate an intracranial tumor by this bystander effect, we injected the tumor cells transduced with the HSV-tk gene (TK cells) in the vicinity of the preimplanted wild-type tumor and then administered GCV. Wild-type 9L-gliosarcoma cells (1 x 10[5]) were implanted into the brain of syngeneic Fisher rats. On the next day, rats were injected with TK cells (1 x 10(5) or 3 x 10[5]) or medium alone at the same brain coordinate and then treated with GCV or saline. Administration of GCV significantly prolonged the survival of the rats injected with TK cells compared with that injected with medium alone (p < 0.01). Reduction in tumor size and retardation of tumor growth were observed by serial magnetic resonance imaging in the rats that received the combination of TK cells and GCV. The results show that the bystander effect is also achieved in vivo even when TK cells and wild-type cells are not simultaneously implanted. This treatment modality circumvents potential risks accompanied with in vivo gene transfer. Because there remained substantially no HSV-tk-positive cells in the recurrent tumors, this modality offers a "safe" therapeutic strategy against human malignant gliomas.


Asunto(s)
Antimetabolitos/farmacología , Neoplasias Encefálicas/terapia , Ganciclovir/farmacología , Terapia Genética/métodos , Gliosarcoma/terapia , Timidina Quinasa/genética , Proteínas Virales/genética , Animales , Neoplasias Encefálicas/diagnóstico por imagen , Técnicas de Transferencia de Gen , Gliosarcoma/diagnóstico por imagen , Imagen por Resonancia Magnética , Masculino , Trasplante de Neoplasias/métodos , Cintigrafía , Ratas , Ratas Endogámicas F344 , Simplexvirus/enzimología , Análisis de Supervivencia , Células Tumorales Cultivadas
16.
J Invest Dermatol ; 115(6): 1065-71, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11121143

RESUMEN

Keloids are benign dermal tumors, characterized by overgrowth of lesions, invasiveness beyond the original boundary of the insult, and recurrence of lesions. The exact etiology is unknown, however. Our hypothesis is that keloids are acquired as a result of an abnormal or prolonged wound healing process, with persistent proliferation and extracellular matrix production of fibroblasts that should otherwise discontinue in normal wound healing. In this study, we examined the response of keloid fibroblasts to proapoptotic signaling. Cell-permeable ceramide, N-acetyl-D-sphingosine, induced apoptosis of dermal fibroblasts in a dose- and time-dependent manner, which was detected by phase contrast microscopy, fluorescent microscopy, the TUNEL method, flow cytometric analysis, and WST-1 assay. In contrast, keloid fibroblasts resisted apoptosis induced by N-acetyl-D-sphingosine (percent survival with 40 mM ceramide treatment for 12 h, normal versus keloid: 9.6% +/- 6.6% vs 66.8% +/- 5.5%). Western blotting analysis showed insulin-like growth factor I receptor overexpression in keloid fibroblasts, but not in normal fibroblasts. Exogenously added insulin-like growth factor I enhanced the resistance of keloid fibroblasts to ceramide-induced apoptosis. Wort- mannin, a phosphatidylinositol 3 kinase inhibitor, suppressed the antiapoptotic action of insulin-like growth factor I in keloid fibroblasts. Our results suggest that keloid fibroblasts overexpressing insulin-like growth factor I receptor are resistant to apoptosis, thus allowing persistent proliferation and production of excessive extracellular matrix. J Invest Dermatol 115:1065-1071 2000


Asunto(s)
Ceramidas/farmacología , Fibroblastos/citología , Queloide/patología , Receptor IGF Tipo 1/biosíntesis , Apoptosis/efectos de los fármacos , Humanos , Fosfatidilinositol 3-Quinasas/farmacología
17.
Endocrinology ; 124(4): 1794-9, 1989 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-2564339

RESUMEN

Insulin-like growth factor I (IGF-I) suppresses GH gene transcription and GH secretion, while somatostatin (SRIF) only suppresses GH secretion. The interaction of these inhibitors of GH was, therefore, tested in primary rat pituitary cells grown in serum-free medium. Maximal inhibition of GH secretion (to 30% of the control value) was achieved by 13 nM IGF-I, while 5 nM SRIF suppressed GH to 36% of control secretion. The respective ED50 values for IGF-I and SRIF inhibition of GH secretion were similar (approximately 2.5 nM). Treatment of cells with the two agents together resulted in a further inhibition of basal GH secretion to 18% of control untreated cells (P less than 0.005). Increasing doses of SRIF (2.5-10 nM) in the presence of IGF-I caused a dose-dependent suppression of GH secretion. PRL levels were not altered by these treatments, indicating a selective effect on GH. GRH-induced GH secretion was further attenuated by combined IGF-I and SRIF treatment compared to the effect of either of these two agents alone. Northern analysis showed that IGF-I suppressed GH mRNA transcripts, while SRIF did not alter GH mRNA levels. The results indicate that physiological concentrations of both IGF-I and SRIF suppress long term basal GH secretion. Only IGF-I alters GH mRNA levels. These two peptides, therefore, appear to attenuate in vitro GH secretion by different mechanisms.


Asunto(s)
Hormona del Crecimiento/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , ARN Mensajero/metabolismo , Somatomedinas/metabolismo , Somatostatina/metabolismo , Animales , Relación Dosis-Respuesta a Droga , Hormona del Crecimiento/genética , Factor I del Crecimiento Similar a la Insulina/farmacología , Masculino , Adenohipófisis/análisis , Adenohipófisis/citología , Adenohipófisis/efectos de los fármacos , ARN Mensajero/análisis , ARN Mensajero/efectos de los fármacos , Ratas , Ratas Endogámicas , Somatostatina/farmacología
18.
Endocrinology ; 132(2): 839-45, 1993 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-8381075

RESUMEN

Although thyrotropin is known to regulate thyroid cell differentiation and proliferation, human thyroid carcinoma cells are relatively insensitive or resistant to TSH stimulation. The expression levels of TSH receptor are significantly lower in carcinoma tissues than in normal tissues. Furthermore, in vitro human thyroid cell growth is not regulated by TSH itself. We, therefore, isolated neomycin-resistant stable human thyroid carcinoma cell (WRO cell) transfectants overexpressing intact human TSH receptor to evaluate the functional role of TSH receptor on carcinoma cells. Southern blot analysis confirmed incorporation and amplification of human TSH receptor complementary DNA sequences into genomic DNA. Northern gel analysis and reverse transcriptase-polymerase chain reaction analysis revealed the presence of specific TSH receptor messenger RNA (4.0 kilobases), and the specific binding and the affinity of [125I]TSH on stably transfected WRO cells were demonstrated compared to wild type. Nevertheless, impaired cAMP production to transfectants by TSH was observed. cAMP production was confirmed after stimulation of both wild type and transfectants by forskolin, cholera toxin, and isoproterenol. In contrast, TSH could affect the cytoplasmic calcium mobilization immediately after the addition of TSH to WRO transfectants. These results suggest that the impairment of TSH action on human thyroid carcinoma cells is not due to a major structural abnormality of the TSH receptor, reduction in the receptor number, or receptor affinity, but much more likely due to a TSH receptor-guanyl nucleotide-binding protein coupling defect.


Asunto(s)
Calcio/metabolismo , AMP Cíclico/metabolismo , Replicación del ADN/efectos de los fármacos , Receptores de Tirotropina/metabolismo , Tirotropina/metabolismo , Tirotropina/farmacología , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Northern Blotting , Southern Blotting , Toxina del Cólera/farmacología , Colforsina/farmacología , Humanos , Isoproterenol/farmacología , Cinética , ARN Mensajero/metabolismo , Receptores de Tirotropina/genética , Proteínas Recombinantes/metabolismo , Neoplasias de la Tiroides , Factores de Tiempo , Transfección , Células Tumorales Cultivadas
19.
Endocrinology ; 138(11): 4577-83, 1997 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9348181

RESUMEN

In an attempt to develop gene therapy for thyroid carcinomas, the present studies were undertaken to evaluate in vitro and in vivo therapeutic efficacy and toxicity of herpes simplex virus thymidine kinase (HSV-tk) gene and ganciclovir (GCV) treatment, a widely used prodrug/suicide gene therapy, in human thyroid carcinoma cell lines, FRO and WRO cells, using a means of retrovirus-mediated gene transduction. In vitro experiments demonstrated dose- and time-dependent cell killing by transduction of the HSV-tk gene followed by GCV treatment. The IC50 (the concentration required to elicit 50% growth inhibition) shifted from 250 to 0.5 mg/liter in FRO cells, and from 3,000 to 0.09 mg/liter in WRO cells with therapeutic indexes of 500 and 33,000, respectively. Treatment with 30 mg/liter GCV for 4 days led to complete cell death in HSV-tk tumor cells. Nontransduced cells mixed with transduced cells were also effectively killed by GCV (bystander effect). Low concentrations of GCV, which alone showed little cytotoxicity, enhanced radiation-induced cytotoxicity (radiosensitization). In vivo sc FRO-tk tumor models in nude mice also showed dose- and time-dependent tumor regression. The IC50 was less than 2 mg/kg, and treatment with 100 mg/kg GCV for 2 weeks completely eradicated all tumors. The bystander effect and radiosensitization were also obtained in vivo. These results suggest that the HSV-tk/GCV approach to human thyroid carcinoma cells appears to be very efficacious, with a wide therapeutic range, and exerts a bystander effect and radiosensitization both in vitro and in vivo. Thus, HSV-tk/GCV system, alone or in combination with radiotherapy, may be a promising suicide gene therapy for thyroid carcinomas.


Asunto(s)
Antivirales/uso terapéutico , Ganciclovir/uso terapéutico , Profármacos/uso terapéutico , Fármacos Sensibilizantes a Radiaciones/uso terapéutico , Simplexvirus/enzimología , Timidina Quinasa/genética , Neoplasias de la Tiroides/radioterapia , Neoplasias de la Tiroides/terapia , Transducción Genética , Adenocarcinoma Folicular/patología , Animales , Carcinoma/patología , Relación Dosis-Respuesta a Droga , Terapia Genética , Vectores Genéticos , Humanos , Ratones , Ratones Desnudos , Retroviridae/genética , Neoplasias de la Tiroides/genética , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/efectos de la radiación
20.
Endocrinology ; 139(2): 803-6, 1998 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9449658

RESUMEN

We here show that the epitope-tagged human TSH receptor (TSHRmyc) is covalently modified with palmitic acid by thioesterification. Side-directed mutagenesis identified Cys699 in the C-terminal cytoplasmic tail of the receptor as the putative palmitoylation site. Mutation of Cys699 to Ala results in the nonpalmitoylated receptor (TSHRmycC699A) in which high affinity TSH binding, Gs coupling, homologous desensitization and TSH-induced internalization are unaffected. In contrast, abolition of palmitoylation appears to decrease the rate of the intracellular trafficking of the receptor. However, since most of TSHRmycC699A seems to be fully processed finally and the receptor number of TSHRmycC699A on the cell surface is comparable to that of TSHRmyc, our results suggest that abolition of palmitoylation delays the cell surface expression of TSHR, but does not trap the receptor intracellularly, although another possibility for proteolytic degradation of either the 95 kDa or the 100 kDa mutant receptor can not be excluded. Thus, post-translational modification of TSHR by palmitoylation may provide a novel mechanism of enhancing the rate of intracellular trafficking of the receptor.


Asunto(s)
Mutación/fisiología , Ácido Palmítico/metabolismo , Receptores de Tirotropina/metabolismo , Secuencia de Aminoácidos , Humanos , Membranas Intracelulares/metabolismo , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Receptores de Tirotropina/genética , Lugares Marcados de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA