Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
J Cell Biol ; 127(1): 203-23, 1994 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-7929563

RESUMEN

Basic fibroblast growth factor (bFGF), a potent mitogenic/neurotrophic factor, controls the development and plasticity of many types of neural cells. In adrenal chromaffin cells, the appearance of bFGF protein coincided with the establishment of functional innervation, suggesting induction by trans-synaptic signals. In cultured bovine adrenal medullary cells Western blot analysis revealed 18-, 23-, and 24-kD bFGF isoforms in the cytosolic and nuclear fractions. Stimulation of acetylcholine nicotinic receptors or hormonal angiotensin II receptors or the direct stimulation of adenylate cyclase with forskolin or protein kinase C (PKC) with PMA increased the content of all bFGF isoforms. Increases in the levels of intracellular bFGF did not result in detectable presence of bFGF proteins in culture medium. Instead, bFGF proteins accumulated in the cytoplasm or the nucleus depending on whether PKC or cAMP pathways were activated. The long-term nuclear forskolin-induced accumulation of bFGF was prevented by cycloheximide or by antisense bFGF oligonucleotide and was also accompanied by an increase in bFGF mRNA. We used luciferase reporter plasmids containing the human bFGF promoter to show that the induction of bFGF resulted from transcriptional activation of the bFGF gene and was mediated by regulatory sequences located upstream from its transcription start site. Stimulation of bFGF gene expression by forskolin and PMA was synergistic and was mediated through different promoter regions. The results suggest that stimulation by cAMP and PKC is mediated through novel cis elements. The regulation of bFGF protein content also involves posttranscriptional mechanisms since changes in the levels of individual bFGF isoforms were different depending on whether cells were treated with carbachol or angiotensin II, forskolin, or PMA. The present study indicates that bFGF is an intracrine cytoplasmic-nuclear factor, whose expression is regulated by trans-synaptic and hormonal stimuli and which may act as a direct mediator of genomic responses to afferent stimulation.


Asunto(s)
Médula Suprarrenal/metabolismo , Factor 2 de Crecimiento de Fibroblastos/análisis , Factor 2 de Crecimiento de Fibroblastos/genética , Regulación de la Expresión Génica , Médula Suprarrenal/citología , Animales , Secuencia de Bases , Bovinos , Núcleo Celular/química , Células Cultivadas , Colforsina/farmacología , AMP Cíclico/farmacología , Citoplasma/química , Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Modelos Biológicos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/genética , Proteína Quinasa C/farmacología , Receptores de Angiotensina/fisiología , Receptores Colinérgicos/fisiología , Transducción de Señal/fisiología , Acetato de Tetradecanoilforbol/farmacología , Activación Transcripcional
2.
Results Probl Cell Differ ; 66: 183-206, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30209660

RESUMEN

Stem cell-derived brain organoids replicate important stages of the prenatal human brain development and combined with the induced pluripotent stem cell (iPSC) technology offer an unprecedented model for investigating human neurological diseases including autism and microcephaly. We describe the history and birth of organoids and their application, focusing on cerebral organoids derived from embryonic stem cells and iPSCs. We discuss new insights into organoid-based model of schizophrenia and shed light on challenges and future applications of organoid-based disease model system. This review also suggests hitherto unrevealed potential applications of organoids in combining with new technologies such as nanophotonics/optogenomics for controlling brain development and atomic force microscopy for studying mechanical forces that shape the developing brain.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Modelos Biológicos , Organoides , Esquizofrenia/patología , Encéfalo/citología , Células Madre Embrionarias/citología , Humanos , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología , Organoides/citología , Organoides/crecimiento & desarrollo
3.
J Neurosci Methods ; 165(2): 230-43, 2007 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-17655935

RESUMEN

Studies have shown the presence of expanded polyQ containing proteins in brain cells related to Huntington disease (HD) and other poly-glutamine disorders. We report the use of organically modified silica (ORMOSIL) nanoparticles as an efficient non-viral gene carrier in an effort to model brain pathology associated with those disorders induced by expanded polyQ peptides. In experiment 1, plasmids expressing Hemaglutinin-tagged polypeptides with 20 glutamine repeats (Q20) or with extended 127-glutamine repeats (Q127) were complexed with ORMOSIL nanoparticles and injected twice (2 weeks apart) into the lateral ventricle of the mouse brain. Fourteen days post-injection of Q127, immunocytochemistry revealed the presence of the characteristic nuclear and cytoplasmic Q127 aggregates in numerous striatal, septal and neocortical neuronal cells as well as ubiquitin-containing aggregates indicative of the neuronal pathology. The mice receiving Q127 showed a marked increase in the reactive GFAP (+) astrocytes in striatum, septum and brain cortex, further indicating the neurodegenerative changes, accompanied by motor impairments. In experiment 2, plasmids Q20 or Q127 were complexed with ORMOSIL and were injected into the brain lateral ventricle or directly into the striatum of adult rats. In both routes of transfection, Q127 induced the appearance of reactive GFAP (+) astrocytes and activated ED1 antigen expressing microglia. An increase in the size of the lateral ventricle was also observed in rats receiving Q127. In transgenic mouse polyQ models, extensive pathologies occur outside the nervous system and the observed brain pathologies could reflect developmental effects of the toxic polyQ proteins. Our experiments show that the nervous tissue restricted expression of poly Q-extended peptides in adult brain is sufficient to evoke neuropathologies associated with HD and other polyQ disorders. Thus, nanotechnology can be employed to model pathological and behavioral aspects of genetic brain diseases in mice as well as in other species, providing a novel research tool for in vivo testing of single or multi-gene therapies.


Asunto(s)
Técnicas de Transferencia de Gen/tendencias , Vectores Genéticos/genética , Nanopartículas/química , Péptidos/genética , Siloxanos/farmacología , Transfección/métodos , Animales , Encéfalo/metabolismo , Encéfalo/patología , Encéfalo/fisiopatología , Expansión de las Repeticiones de ADN/genética , Modelos Animales de Enfermedad , Ectodisplasinas/análisis , Ectodisplasinas/biosíntesis , Femenino , Gliosis/genética , Gliosis/metabolismo , Gliosis/fisiopatología , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/fisiopatología , Inyecciones Intraventriculares , Masculino , Ratones , Ratones Transgénicos , Nanopartículas/toxicidad , Degeneración Nerviosa/genética , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Péptidos/metabolismo , Péptidos/toxicidad , Plásmidos/genética , Ratas , Ratas Wistar , Dióxido de Silicio
4.
Mol Biol Cell ; 7(8): 1299-317, 1996 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-8856671

RESUMEN

In an effort to determine the localization of fibroblast growth factor (FGF) receptors (FGFR) that could mediate the intracellular action of FGF-2, we discovered the presence of high-affinity. FGF-2 binding sites in the nuclei of bovine adrenal medullary cells (BAMC). Western blot analysis demonstrated the presence of 103-, 118-, and 145-kDa forms of FGFR1 in nuclei isolated from BAMC. 125I-FGF-2 cross-linking to nuclear extracts followed by FGFR1 immunoprecipitation showed that FGFR1 can account for the nuclear FGF-2 binding sites. Nuclear FGFR1 has kinase activity and undergoes autophosphorylation. Immunocytochemistry with the use of confocal and electron microscopes demonstrated the presence of FGFR1 within the nuclear interior. Nuclear subfractionation followed by Western blot or immunoelectron microscopic analysis showed that the nuclear FGFR1 is contained in the nuclear matrix and the nucleoplasm. Agents that induce translocation of endogenous FGF-2 to the nucleus (forskolin, carbachol, or angiotensin II) increased the intranuclear accumulation of FGFR1. This accumulation was accompanied by an overall increase in FGF-2-inducible tyrosine kinase activity. Our findings suggest a novel mode for growth factor action whereby growth factor receptors translocate to the nucleus in parallel with their ligand and act as direct mediators of nuclear responses to cell stimulation.


Asunto(s)
Médula Suprarrenal/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Médula Suprarrenal/citología , Animales , Bovinos , Núcleo Celular/metabolismo , Células Cultivadas , Cinética , Microscopía Inmunoelectrónica , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal , Fracciones Subcelulares/metabolismo
5.
Mol Biol Cell ; 9(8): 2269-85, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9693381

RESUMEN

Induction of the fibroblast growth factor-2 (FGF-2) gene and the consequent accumulation of FGF-2 in the nucleus are operative events in mitotic activation and hypertrophy of human astrocytes. In the brain, these events are associated with cellular degeneration and may reflect release of the FGF-2 gene from cell contact inhibition. We used cultures of human astrocytes to examine whether expression of FGF-2 is also controlled by soluble growth factors. Treatment of subconfluent astrocytes with interleukin-1beta, epidermal or platelet-derived growth factors, 18-kDa FGF-2, or serum or direct stimulation of protein kinase C (PKC) with phorbol 12-myristate 13-acetate or adenylate cyclase with forskolin increased the levels of 18-, 22-, and 24-kDa FGF-2 isoforms and FGF-2 mRNA. Transfection of FGF-2 promoter-luciferase constructs identified a unique -555/-513 bp growth factor-responsive element (GFRE) that confers high basal promoter activity and activation by growth factors to a downstream promoter region. It also identified a separate region (-624/-556 bp) essential for PKC and cAMP stimulation. DNA-protein binding assays indicated that novel cis-acting elements and trans-acting factors mediate activation of the FGF-2 gene. Southwestern analysis identified 40-, 50-, 60-, and 100-kDa GFRE-binding proteins and 165-, 112-, and 90-kDa proteins that interacted with the PKC/cAMP-responsive region. The GFRE and the element essential for PKC and cAMP stimulation overlap with the region that mediates cell contact inhibition of the FGF-2 promoter. The results show a two-stage regulation of the FGF-2 gene: 1) an initial induction by reduced cell contact, and 2) further activation by growth factors or the PKC-signaling pathway. The hierarchic regulation of the FGF-2 gene promoter by cell density and growth factors or PKC reflects a two-stage activation of protein binding to the GFRE and to the PKC/cAMP-responsive region, respectively.


Asunto(s)
Astrocitos/metabolismo , Factor 2 de Crecimiento de Fibroblastos/genética , Regulación de la Expresión Génica , Plasticidad Neuronal/fisiología , Regiones Promotoras Genéticas , Transcripción Genética , Astrocitos/citología , Astrocitos/fisiología , Secuencia de Bases , Sitios de Unión , Encéfalo/citología , Encéfalo/metabolismo , Núcleo Celular/metabolismo , Células Cultivadas , Colforsina/farmacología , Proteínas de Unión al ADN/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Factor 2 de Crecimiento de Fibroblastos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-1/farmacología , Luciferasas/biosíntesis , Mitosis , Datos de Secuencia Molecular , Oligodesoxirribonucleótidos/farmacología , Oligonucleótidos Antisentido/farmacología , Reacción en Cadena de la Polimerasa , Proteínas Recombinantes de Fusión/biosíntesis , Acetato de Tetradecanoilforbol/farmacología , Transcripción Genética/efectos de los fármacos , Transfección
6.
Mol Biol Cell ; 12(2): 449-62, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11179427

RESUMEN

In bovine adrenal medullary cells synergistically acting type 1 and type 2 angiotensin II (AII) receptors activate the fibroblast growth factor-2 (FGF-2) gene through a unique AII-responsive promoter element. Both the type 1 and type 2 AII receptors and the downstream cyclic adenosine 1',3'-monophosphate- and protein kinase C-dependent signaling pathways activate the FGF-2 promoter through a novel signal-transducing mechanism. This mechanism, which we have named integrative nuclear FGF receptor-1 signaling, involves the nuclear translocation of FGF receptor-1 and its subsequent transactivation of the AII-responsive element in the FGF-2 promoter.


Asunto(s)
Núcleo Celular/metabolismo , Factor 2 de Crecimiento de Fibroblastos/genética , Receptores de Angiotensina/metabolismo , Transducción de Señal , Médula Suprarrenal/citología , Médula Suprarrenal/metabolismo , Angiotensina II/análogos & derivados , Angiotensina II/farmacología , Animales , Sitios de Unión , Bovinos , Núcleo Celular/genética , Células Cultivadas , Huella de ADN , Desoxirribonucleasa I/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Imidazoles/farmacología , Regiones Promotoras Genéticas , Piridinas/farmacología , Receptor de Angiotensina Tipo 1 , Receptor de Angiotensina Tipo 2 , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Elementos de Respuesta/fisiología , Transactivadores/metabolismo , Tirosina/metabolismo
7.
Schizophr Res ; 185: 17-32, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28094170

RESUMEN

The watershed-hypothesis of schizophrenia asserts that over 200 different mutations dysregulate distinct pathways that converge on an unspecified common mechanism(s) that controls disease ontogeny. Consistent with this hypothesis, our RNA-sequencing of neuron committed cells (NCCs) differentiated from established iPSCs of 4 schizophrenia patients and 4 control subjects uncovered a dysregulated transcriptome of 1349 mRNAs common to all patients. Data reveals a global dysregulation of developmental genome, deconstruction of coordinated mRNA networks, and the formation of aberrant, new coordinated mRNA networks indicating a concerted action of the responsible factor(s). Sequencing of miRNA transcriptomes demonstrated an overexpression of 16 miRNAs and deconstruction of interactive miRNA-mRNA networks in schizophrenia NCCs. ChiPseq revealed that the nuclear (n) form of FGFR1, a pan-ontogenic regulator, is overexpressed in schizophrenia NCCs and overtargets dysregulated mRNA and miRNA genes. The nFGFR1 targeted 54% of all human gene promoters and 84.4% of schizophrenia dysregulated genes. The upregulated genes reside within major developmental pathways that control neurogenesis and neuron formation, whereas downregulated genes are involved in oligodendrogenesis. Our results indicate (i) an early (preneuronal) genomic etiology of schizophrenia, (ii) dysregulated genes and new coordinated gene networks are common to unrelated cases of schizophrenia, (iii) gene dysregulations are accompanied by increased nFGFR1-genome interactions, and (iv) modeling of increased nFGFR1 by an overexpression of a nFGFR1 lead to up or downregulation of selected genes as observed in schizophrenia NCCs. Together our results designate nFGFR1 signaling as a potential common dysregulated mechanism in investigated patients and potential therapeutic target in schizophrenia.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Genoma/genética , Mutación/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Esquizofrenia/genética , Transducción de Señal/genética , Adulto , Diferenciación Celular , Células Cultivadas , Femenino , Redes Reguladoras de Genes , Genómica , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Modelos Biológicos , Receptor Notch1/metabolismo , Esquizofrenia/patología , Transcriptoma , Adulto Joven
8.
Transl Psychiatry ; 7(11): 6, 2017 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-30446636

RESUMEN

Studies of induced pluripotent stem cells (iPSCs) from schizophrenia patients and control individuals revealed that the disorder is programmed at the preneuronal stage, involves a common dysregulated mRNA transcriptome, and identified Integrative Nuclear FGFR1 Signaling a common dysregulated mechanism. We used human embryonic stem cell (hESC) and iPSC-derived cerebral organoids from four controls and three schizophrenia patients to model the first trimester of in utero brain development. The schizophrenia organoids revealed an abnormal scattering of proliferating Ki67+ neural progenitor cells (NPCs) from the ventricular zone (VZ), throughout the intermediate (IZ) and cortical (CZ) zones. TBR1 pioneer neurons and reelin, which guides cortico-petal migration, were restricted from the schizophrenia cortex. The maturing neurons were abundantly developed in the subcortical regions, but were depleted from the schizophrenia cortex. The decreased intracortical connectivity was denoted by changes in the orientation and morphology of calretinin interneurons. In schizophrenia organoids, nuclear (n)FGFR1 was abundantly expressed by developing subcortical cells, but was depleted from the neuronal committed cells (NCCs) of the CZ. Transfection of dominant negative and constitutively active nFGFR1 caused widespread disruption of the neuro-ontogenic gene networks in hESC-derived NPCs and NCCs. The fgfr1 gene was the most prominent FGFR gene expressed in NPCs and NCCs, and blocking with PD173074 reproduced both the loss of nFGFR1 and cortical neuronal maturation in hESC cerebral organoids. We report for the first time, progression of the cortical malformation in schizophrenia and link it to altered FGFR1 signaling. Targeting INFS may offer a preventive treatment of schizophrenia.


Asunto(s)
Corteza Cerebral/patología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Esquizofrenia/patología , Calbindina 2/metabolismo , Corteza Cerebral/metabolismo , Células Madre Embrionarias/patología , Humanos , Interneuronas/metabolismo , Interneuronas/patología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Proteína Reelina , Esquizofrenia/genética , Esquizofrenia/metabolismo
9.
Oncogene ; 14(18): 2201-11, 1997 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-9174056

RESUMEN

In this study we describe the presence of high affinity FGF-2 binding sites in the nuclei of U251MG glioma cells (K(d)=7 pM). Immunoprecipitation of total cell extracts with FGF receptor (FGFR) 1-4 antibodies showed that U251MG glioma cells express only FGFR1. [125I]FGF-2 cross linking to nuclear extracts followed by FGFR1 immunoprecipitation showed that FGFR1 may account for the nuclear FGF-2 binding sites. Western blot analysis demonstrated the presence of 103, 118 kDa and small amounts of 145 kDa FGFR1 isoforms in the nuclei of glioma cells. All isoforms contain both the C- and N-terminal domains. Nuclear FGFR1 retains kinase activity. Immunocytochemistry using confocal microscopy showed specific FGFR1 immunoreactivity within the nuclear interior. In continuously proliferating glioma cells, nuclear FGFR1 is constitutively expressed, independent of cell density. In contrast, in nontransformed human astrocytes, nuclear FGFR1 levels fluctuate with the proliferative state of the cell. In quiescent, confluent astrocytes nuclear FGFR1 protein was depleted. An accumulation of nuclear FGFR1 was observed following the transition to a subconfluent, proliferating state. Transfection of a pcDNA3.1-FGFR1 expression vector into glioma cells that do not express FGFR1 resulted in the nuclear accumulation of FGFR1, increased cell proliferation, and stimulated transition from the G0/G1 to the S-phase of the cell cycle. The increased proliferative rate was resistant to inhibition by the cell-impermeable FGF binding antagonist, myoinositol hexakis [dihydrogen phosphate]. Our results suggest that the constitutive nuclear presence of FGFR1 contributes to the increased proliferation of glioma cells while the transient nuclear accumulation of FGFR1 in normal astrocytes may play a role in the transition to a reactive state.


Asunto(s)
Núcleo Celular/metabolismo , Glioma/metabolismo , Neuroglía/citología , Neuroglía/metabolismo , Proteínas Tirosina Quinasas Receptoras , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Astrocitos/química , Astrocitos/metabolismo , Western Blotting , División Celular/fisiología , Núcleo Celular/química , Células Cultivadas , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Glioma/química , Humanos , Inmunohistoquímica , Radioisótopos de Yodo , Mitógenos/fisiología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Receptores Citoplasmáticos y Nucleares/química , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores de Factores de Crecimiento de Fibroblastos/química , Receptores de Factores de Crecimiento de Fibroblastos/inmunología , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Fracciones Subcelulares , Transfección
10.
Oncogene ; 14(2): 171-83, 1997 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-9010219

RESUMEN

FGF-2 has been implicated in the neoplastic transformation of glioma cells and in the transition of normal quiescent astrocytes to a proliferating, reactive state. In the present study we have observed that in human glial cells, levels and subcellular localization of FGF-2 are different in quiescent and proliferating cells. FGF-2 was detected in the cytoplasm of non-reactive astrocytes in human brain sections. In contrast FGF-2 was located within the cytoplasm and nuclei of reactive astrocytes in gliotic brain tissue and in neoplastic cells of glioma tumors. In vitro, FGF-2 was found predominantly in the nucleus of subconfluent proliferating astrocytes, but was detected only in the cytoplasm of density arrested quiescent astrocytes. Our results suggest that reduced cell contact stimulates nuclear accumulation of FGF-2, accompanying mitotic activation of reactive human astrocytes. FGF-2 was constitutively localized to the nucleus of continuously proliferating glioma cells independent of cell density. A role for intracellular FGF-2 was further suggested by the observation that glioma cells that are not stimulated to proliferate by extracellular FGF-2 proliferated faster when transfected with FGF-2 expressing vectors. This increased proliferation correlated with nuclear accumulation of FGF-2. Cell proliferation was attenuated by 5'-deoxy-5'-methylthioadenosine, a FGF-2 receptor tyrosine kinase inhibitor that acts within the cell, but was unaffected by myo-inositol hexakis [dihydrogen phosphate] that disrupts FGF-2 binding to plasma membrane receptors. Our results indicate that FGF-2 serves as a nuclear regulator of proliferation in astrocytic cells. In glioma cells, the constitutive presence of FGF-2 in the nucleus may promote proliferation that is insensitive to cell contact inhibition.


Asunto(s)
Astrocitos/citología , Núcleo Celular/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Glioblastoma/patología , Astrocitos/metabolismo , Comunicación Celular , Recuento de Células , División Celular , Células Cultivadas , Femenino , Glioblastoma/metabolismo , Humanos , Masculino , Neuroglía/citología , Neuroglía/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo
11.
Oncogene ; 5(1): 69-73, 1990 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-2108403

RESUMEN

The nuclear proto-oncogene, c-fos, has been implicated in the coordinated regulation of gene expression during cell proliferation and differentiation. In this study, we have demonstrated the induction of the c-fos gene products in differentiated cells of the adrenal medulla by non-mitogenic signals. Activation of adrenal medullary cells in vivo by insulin-induced hypoglycemia, and in vitro by nicotine or angiotensin resulted in the rapid and transient elevation of c-fos mRNA levels. Induction of the c-fos mRNA by angiotensin and nicotine were accompanied by the appearance of the c-fos protein. The increase in c-fos protein occurred initially in the cytoplasm and, later, in the nucleus, and it was co-localized with tyrosine hydroxylase. Nuclear expression of the c-fos protein was also induced by veratridine, forskolin and the calcium ionophore A231287. The role of calcium in the regulation of the c-fos gene by angiotensin with nifedipine and inhibition of the effects of angiotensin with nifedipine and sphingosine, a protein kinase C inhibitor. Activation of the c-fos gene may play a role in the coordinated induction of genes involved in the long-term adaptation of adrenal medullary cells to increased functional demands.


Asunto(s)
Médula Suprarrenal/metabolismo , Proteínas Proto-Oncogénicas/genética , Proto-Oncogenes , Angiotensinas/farmacología , Animales , Calcio/fisiología , Células Cultivadas , Expresión Génica , Masculino , Nicotina/farmacología , Proteína Quinasa C/fisiología , Proteínas Proto-Oncogénicas/análisis , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-fos , ARN Mensajero/análisis , Ratas , Ratas Endogámicas F344
12.
Folia Morphol (Warsz) ; 64(4): 273-81, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16425153

RESUMEN

This study shows that an ICP4-replication-deficient herpes simplex virus containing the Moloney murine leukaemia virus LTR fused with the coding sequence for the beta-galactosidase gene can be used as a very effective vector for delivering the beta-galactosidase reporter gene into the rat brain septum. F344 rats received bilateral stereotaxic injections into the nucleus of the diagonal band and into the medial septum. The X-gal stain was used to detect the activity of the expressed beta-galactosidase enzyme. The delivered reporter gene was expressed successfully not only in the neuronal cells of the injected areas but also in cells that project to the injection area such as cortex cells about 6 mm away from the injection sites. Expression was visible at 1, 3 and 9 weeks following injection. We conclude that this vector can effectively deliver genes into different regions of the mature mammalian brain and also to areas distant from the injection site.


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos , Herpesvirus Humano 1 , Núcleos Septales , Animales , Genes Reporteros , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/metabolismo , Humanos , Microinyecciones , Distribución Aleatoria , Ratas , Ratas Endogámicas F344 , Núcleos Septales/anatomía & histología , Núcleos Septales/fisiología , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
13.
Mol Neurobiol ; 15(3): 257-83, 1997 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9457702

RESUMEN

This article discusses a novel intracrine mechanism of growth-factor action in the nervous system whereby fibroblast growth factor-2 (FGF-2) and its receptor accumulate in the cell nucleus and act as mediators in the control of cell growth and proliferation. In human and rat brain the levels and subcellular localization of FGF-2 differ between quiescent and reactive astrocytes. Quiescent cells express a low level of FGF-2, which is located predominantly within the cytoplasm. In reactive astrocytes, the expression of FGF-2 increases and the proteins are found in both the cytoplasm and nucleus. In glioma tumors, FGF-2 is overexpressed in the nuclei of neoplastic cells. Similar changes in FGF-2 expression and localization are found in vitro. The nuclear accumulation of FGF-2 reflects a transient activation of the FGF-2 gene by potentially novel transactivating factors interacting with an upstream regulatory promoter region. In parallel with FGF-2, the nuclei of astrocytes contain the high-affinity FGF-2 receptor, FGFR1. Nuclear FGFR1 is full length, retains kinase activity, and is localized within the nuclear interior in association with the nuclear matrix. Transfection of either FGF-2 or FGFR1 into cells that do not normally express these proteins results in their nuclear accumulation and concomitant increases in cell proliferation. A similar regulation of nuclear FGF-2 and FGFR1 is observed in neural crestderived adrenal medullary cells and of FGF-2 in the nuclei of cerebellar neurons. Thus, the regulation of the nuclear content of FGF-2 and FGFR1 could serve as a novel mechanism controlling growth and proliferation of glial and neuronal cells.


Asunto(s)
Astrocitos/citología , Factor 2 de Crecimiento de Fibroblastos/fisiología , Proteínas Tirosina Quinasas Receptoras , Animales , División Celular , Humanos , Neuroglía/citología , Proteína Quinasa C/fisiología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Transducción de Señal , Células Tumorales Cultivadas
14.
Arch Neurol ; 41(8): 856-61, 1984 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-6147127

RESUMEN

Parkinson's disease usually involves a lengthy preclinical period during which few neurological symptoms are observed despite extensive damage to the dopaminergic nigrostriatal bundle. Injury to this projection in the rat also fails to produce major neurological dysfunctions. In our studies, damage to the nigrostriatal bundle of the rat, resulting in the loss of up to 95% of the dopaminergic terminals in striatum, was accompanied by apparent increases in the synthesis and release of dopamine (DA) from those dopaminergic terminals that remained. More specifically, both the activity of the rate-limiting biosynthetic enzyme, tyrosine hydroxylase, and the content of the principal DA metabolite, dihydroxyphenylacetic acid, were increased in striatum relative to DA levels. The increases were exponentially related to DA loss.


Asunto(s)
Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Enfermedad de Parkinson/metabolismo , Tirosina 3-Monooxigenasa/metabolismo , Animales , Hidroxidopaminas/farmacología , Masculino , Oxidopamina , Ratas , Ratas Endogámicas
15.
BMC Neurosci ; 2: 2, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11231879

RESUMEN

BACKGROUND: Chemical methods of transfection that have proven successful with cell lines often do not work with primary cultures of neurons. Recent data, however, suggest that linear polymers of the cation polyethyleneimine (PEI) can facilitate the uptake of nucleic acids by neurons. Consequently, we examined the ability of a commercial PEI preparation to allow the introduction of foreign genes into postmitotic mammalian neurons. Sympathetic neurons were obtained from perinatal rat pups and maintained for 5 days in vitro in the absence of nonneuronal cells. Cultures were then transfected with varying amounts of a plasmid encoding either E. coli beta-galactosidase or enhanced green fluorescence protein (EGFP) using PEI. RESULTS: Optimal transfection efficiency was observed with 1 microg/ml of plasmid DNA and 5 microg/ml PEI. Expression of beta-galactosidase was both rapid and stable, beginning within 6 hours and lasting for at least 21 days. A maximum yield was obtained within 72 hours with approximately 9% of the neurons expressing beta-galactosidase, as assessed by both histochemistry and antibody staining. Cotransfection of two plasmids encoding reporter genes was achieved. Postmitotic neurons from adult human retinal cultures also demonstrated an ability to take up and express foreign DNA using PEI as a vector. CONCLUSIONS: These data suggest that PEI is a useful agent for the stable expression of plasmid-encoded genes in neuronal cultures.


Asunto(s)
Ganglios Simpáticos/metabolismo , Neuronas/metabolismo , Polietileneimina/metabolismo , Retina/metabolismo , Transfección/métodos , Animales , Animales Recién Nacidos , Células Cultivadas , ADN/metabolismo , Relación Dosis-Respuesta a Droga , Ganglios Simpáticos/citología , Expresión Génica , Humanos , Proteínas Luminiscentes/biosíntesis , Proteínas Luminiscentes/genética , Mitosis , Neuronas/citología , Neuronas/efectos de los fármacos , Plásmidos/genética , Polietileneimina/farmacología , Ratas , Retina/citología , beta-Galactosidasa/biosíntesis , beta-Galactosidasa/genética
16.
Biochem Pharmacol ; 35(8): 1241-8, 1986 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-2421731

RESUMEN

Adult rats received intraventricular injections of 5,7-dihydroxytryptamine (5,7-DHT) to destroy serotonin (5-HT)-containing nerve terminals throughout the brain. When the animals were killed 3 or 21 days later, we observed a marked decrease in 5-HT content in septum and hippocampus and a parallel decline in in vitro high affinity 5-HT uptake. 5-Hydroxyindoleacetic acid (5-HIAA) concentrations also were reduced but by a much smaller extent, resulting in significant increases in the ratio of 5-HIAA to 5-HT. These changes were accompanied by similar increases in the ratio of tryptophan hydroxylase (TPH) activity to 5-HT content. The relative increases in TPH activity resulted from two temporally distinct processes, the first of which appeared to be an activation that could be mimicked in vitro by Ca2+-dependent phosphorylation. We conclude that, after partial damage to 5-HT neurons, there is a compensatory increase in the synthesis and release of 5-HT from those terminals that remain.


Asunto(s)
5,7-Dihidroxitriptamina/farmacología , Dihidroxitriptaminas/farmacología , Receptores de Serotonina/metabolismo , Triptófano Hidroxilasa/análisis , Animales , Encéfalo/metabolismo , Retroalimentación , Ácido Hidroxiindolacético/análisis , Técnicas In Vitro , Cinética , Masculino , Núcleos del Rafe/enzimología , Ratas , Ratas Endogámicas , Serotonina/análisis , Serotonina/metabolismo
17.
Brain Res Mol Brain Res ; 55(2): 293-305, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9582440

RESUMEN

FGF-2, a mitogenic/neurotrophic protein, controls the development and plasticity of many types of neural cells. In neural crest-derived adrenal pheochromatocytes, induction of FGF-2 coincides with the establishment of functional innervation and is reproduced in vitro by stimulating acetylcholine receptors (AChR). The mechanisms by which AChR activate the FGF-2 gene were examined in cultured bovine adrenal medullary chromaffin (BAMC) cells in which AChR induce expression and nuclear accumulation of growth-promoting FGF-2 and FGF-2 receptors. Carbachol or nicotine increased expression of transfected FGF-2 gene promoter-luciferase constructs and were more potent than the muscarinic agonist ABMCB. Deletion analysis has identified a unique -555/-512 bp element that confers AChR stimulation and basal activity to the downstream FGF-2 promoter, and a separate protein kinase C/cAMP-responsive sequence (-625/-555 bp). Stimulation of AChR increased in vitro formation of protein complexes with the AChR-responsive element which were not displaced by target oligonucleotides for common trans-activators. Southwestern analysis identified 50-55, 125, 140 and 170 kDa proteins that interact with the AChR-responsive element in a manner stimulated by AChR. Nicotine increased tyrosine phosphorylation of cytoplasmic and nuclear proteins, including 50-55 kDa promoter-binding factors. Activation of the FGF-2 promoter was reduced by genistein. Thus, nicotinic AChR activate the FGF-2 gene via a new signaling mechanism separate from the cAMP/PKC pathways. It utilizes tyrosine phosphorylation and interaction of trans-activating factors with a novel cis-acting element. It offers a new pathway through which trans-synaptic signals may control neural development and plasticity.


Asunto(s)
Regiones no Traducidas 5'/fisiología , Factor 2 de Crecimiento de Fibroblastos/genética , Plasticidad Neuronal/genética , Receptores Nicotínicos/fisiología , Sinapsis/fisiología , Activación Transcripcional , Tirosina/metabolismo , Glándulas Suprarrenales/citología , Animales , Bovinos , Diferenciación Celular/genética , Células Cultivadas , Células Cromafines , AMP Cíclico/fisiología , Fosforilación , Regiones Promotoras Genéticas/fisiología , Unión Proteica/genética , Proteína Quinasa C/fisiología
18.
Brain Res Mol Brain Res ; 36(1): 193-6, 1996 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9011759

RESUMEN

We examined expression of the 1,25-dihydroxyvitamin D3 [1,25-(OH)2 D3] receptors in chromaffin cells of the adrenal medulla and the effects of 1,25(OH)2 D3 on expression of the tyrosine hydroxylase (TH) gene. Accumulation of 1,25(OH)2 D3 in the nuclei of adrenal medullary cells, but not in the adrenal cortex, was observed in mice intravenously injected with radioactively labeled hormone. 1,25(OH)2 D3 produced concentration-dependent increases in the TH mRNA levels in cultured bovine adrenal medullary cells (BAMC). The maximal increases (2-3-fold) occurred at 10(-8) M 1,25(OH)2 D3. Combined treatment with 1,25(OH)2 D3 and 20 microM nicotine had no additive effect on TH mRNA levels suggesting that transsynaptic (nicotinic) and vitamin D (hormonal) stimulation of TH gene expression are mediated through converging mechanisms. Induction of TH mRNA by 1,25(OH)2 D3 was not affected by calcium antagonist TMB-8. By increasing expression of the rate limiting enzyme in the catecholamine biosynthetic pathway, 1,25-(OH)2 D3 may participate in the regulation of catecholamine production in adrenal chromaffin cells. This regulation provides mechanisms through which 1,25(OH)2 D3 may control response and adaptation to stress.


Asunto(s)
Médula Suprarrenal/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Receptores de Calcitriol/genética , Sistema Nervioso Simpático/metabolismo , Tirosina 3-Monooxigenasa/genética , Vitamina D/farmacología , Médula Suprarrenal/citología , Médula Suprarrenal/metabolismo , Análisis de Varianza , Animales , Bovinos , Técnicas In Vitro , Masculino , Ratones , Neurotransmisores/genética
19.
Brain Res Mol Brain Res ; 22(1-4): 309-19, 1994 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-7912405

RESUMEN

Activation of the tyrosine hydroxylase (TH) gene in the adrenal medulla during stress is mediated by trans-synaptic mechanisms and may involve cholinergic receptors. Stimulation of nicotinic receptors in adrenal medullary cells induces cell depolarization, influx of Ca2+ ions and increases levels of cAMP. We have shown that both cAMP and membrane depolarization produce an increase in the expression of the TH gene in cultured bovine adrenal medullary cells (BAMC). Others have proposed that transcriptional activation of the TH gene by cAMP is mediated through the sequence homologous to a cAMP responsive element (CRE) located in the proximal region of the TH gene promoter. In the present study we have examined the mechanisms by which membrane depolarization increases the TH gene activity. Treatment of serum-free BAMC cultures with the depolarizing agent, veratridine, increased the extracellular concentration of catecholamines, Met5-enkephalin, and the relative abundance of TH mRNA. Veratridine treatment also increased the levels of mRNAs for the catecholamine biosynthetic enzyme phenylethanolamine N-methyltransferase (PNMT), and proenkephalin A (PEK). Treatment for longer than 3 h was required to increase TH mRNA levels. By contrast, our previous studies indicated that cAMP stimulation for 2 h produces a maximal increase in TH mRNA levels in BAMC. The effects of veratridine and forskolin on TH mRNA levels were additive, further indicating that depolarization and cAMP activate TH gene expression via different pathways. Calmidazolium, an antagonist of calmodulin, had no effect on the veratridine-induced increase in TH mRNA levels. Similarly sphingosine treatment or preincubation with PMA, which reduce protein kinase C (PKC) activity and attenuate the induction of TH mRNA by PMA or the hormone, angiotensin II, did not affect the induction by veratridine. To identify promoter mechanisms of TH gene activation in depolarized cells we transfected BAMC with a plasmid pTHgoodLuc and treated with veratridine for 24 h. pTHgoodLUC contains a luciferase reporter gene linked to a -428/+21 bp fragment of the bovine TH gene promoter (relative to the transcription start site). Veratridine increased the expression of luciferase from the TH promoter 2.5-fold. Deletion of the -194/-54 bp promoter region containing SP-1 and POU/Oct sites reduced veratridine stimulation by 40%. Additional deletion of the -269 to -190 bp promoter segment, including an AP-1 element, further reduced veratridine stimulation to a statistically non-significant level. In conclusion, activation of TH gene expression upon depolarization is not mediated by calmodulin and PKC. Promoter sequences involved in this activation are located upstream from the CRE. Depolarization may activate TH gene transcription by acting on more than one regulatory region.


Asunto(s)
Médula Suprarrenal/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regiones Promotoras Genéticas , Sistemas de Mensajero Secundario/efectos de los fármacos , Tirosina 3-Monooxigenasa/genética , Médula Suprarrenal/citología , Médula Suprarrenal/metabolismo , Animales , Calmodulina/fisiología , Bovinos , Colforsina/farmacología , Encefalinas/genética , Potenciales de la Membrana/efectos de los fármacos , Feniletanolamina N-Metiltransferasa/genética , Proteína Quinasa C/fisiología , Precursores de Proteínas/genética , ARN Mensajero/efectos de los fármacos , Activación Transcripcional , Veratridina/farmacología
20.
Brain Res Mol Brain Res ; 38(1): 161-5, 1996 May.
Artículo en Inglés | MEDLINE | ID: mdl-8737680

RESUMEN

Fractionation of human astrocytes revealed the presence of 103, 118, and 145 kDa forms of FGF receptor 1 (FGFR1) in isolated nuclei. Only trace amounts of FGFR1 proteins were detected in the cell membrane or cytoplasmic fractions. Nuclear FGFR1 is found in the nucleoplasm and nuclear matrix but not in chromatin. Immuno-confocal microscopy further demonstrates the intranuclear presence of FGFR1 and its colocalization with FGF-2. Nuclear FGFR1 binds to FGF-2 and has tyrosine kinase activity. Translocation of functional growth factor receptors into the cell nucleus offers a novel mechanism for growth factor action.


Asunto(s)
Astrocitos/química , Núcleo Celular/química , Factor 1 de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/análisis , Membrana Celular/química , Células Cultivadas , Citoplasma/química , Humanos , Peso Molecular
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda