Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Brain Dev ; 40(5): 367-375, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29429559

RESUMEN

In adult rats, erythropoietin improved outcomes early and late after traumatic brain injury, associated with increased levels of Brain Derived Neurotrophic Factor. Using our model of pediatric traumatic brain injury, controlled cortical impact in 17-day old rats, we previously showed that erythropoietin increased hippocampal neuronal fraction in the first two days after injury. Erythropoietin also decreased activation of caspase3, an apoptotic enzyme modulated by Brain Derived Neurotrophic Factor, and improved Novel Object Recognition testing 14 days after injury. Data on long-term effects of erythropoietin on Brain Derived Neurotrophic Factor expression, histology and cognitive function after developmental traumatic brain injury are lacking. We hypothesized that erythropoietin would increase Brain Derived Neurotrophic Factor and improve long-term object recognition in rat pups after controlled cortical impact, associated with increased neuronal fraction in the hippocampus. METHODS: Rats pups received erythropoietin or vehicle at 1, 24, and 48 h and 7 days after injury or sham surgery followed by histology at 35 days, Novel Object Recognition testing at adulthood, and Brain Derived Neurotrophic Factor measurements early and late after injury. RESULTS: Erythropoietin improved Novel Object Recognition performance and preserved hippocampal volume, but not neuronal fraction, late after injury. CONCLUSIONS: Improved object recognition in erythropoietin treated rats was associated with preserved hippocampal volume late after traumatic brain injury. Erythropoietin is approved to treat various pediatric conditions. Coupled with exciting experimental and clinical studies suggesting it is beneficial after neonatal hypoxic ischemic brain injury, our preliminary findings support further study of erythropoietin use after developmental traumatic brain injury.


Asunto(s)
Lesiones Traumáticas del Encéfalo/terapia , Cognición/efectos de los fármacos , Eritropoyetina/uso terapéutico , Animales , Animales Recién Nacidos , Lesiones Traumáticas del Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Eritropoyetina/metabolismo , Eritropoyetina/farmacología , Hipocampo/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Modelos Animales , Neuronas/metabolismo , Ratas , Ratas Sprague-Dawley
2.
Open Biochem J ; 7: 54-65, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23919090

RESUMEN

We investigated whether a cell-penetrating peptide linked via a disulfide bond to a fluorophore-labeled cargo peptide can be used to interrogate changes in cellular redox state. A fluorescence resonance energy transfer (FRET) pair was constructed so that the cargo peptide was labeled with fluorescein amidite (FAM) and the cell-penetrating peptide was attached to a quencher. Incubation of cells in culture with the FRET construct was visualized using live-cell, time-lapse imaging, which demonstrated earlier cellular uptake of the construct when cells were treated with the reducing agent n-acetylcysteine (NAC). The FRET peptide construct was easily detected in cells cultured in 96-well plates using a plate-reader. Treatment of cells with various classes of reducing or oxidizing agents resulted in an increase or decrease in FAM fluorescence, respectively. Changes in FAM fluorescence correlated significantly with redox-sensitive green fluorescent protein ratios in cells treated with hydrogen peroxide but not NAC. Detection of relative changes in cellular redox state was enhanced by the fact that uptake of the cell-penetrating peptide occurred more quickly in relatively reduced compared with oxidized cells. We conclude that cell-penetrating peptides coupled via disulfide bonds to detectable cargo is a novel and specific approach for assessment of relative changes in cellular thiol redox state.

3.
Anal Chem ; 84(21): 9379-87, 2012 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-23043216

RESUMEN

Current microarray assay technology predominately uses fluorescence as a detectable signal end point. This study assessed real-time in situ surface hybridization capture kinetics for single printed DNA microspots on solid array surfaces using fluorescence. The influence of the DNA target and probe cyanine dye position on oligo-DNA duplex formation behavior was compared in solution versus surface-hybridized single DNA printed spots using fluorescence resonance energy transfer (FRET) analysis. Fluorophore Cy3/Cy5 fluorescence intensities were analyzed both through the printed hybridized DNA spot thickness and radially across single-spot surfaces. Confocal single-spot imaging shows that real-time in situ hybridization kinetics with constant target concentrations changes as a function of the printed probe density. Target-specific imaging in single spots exhibits a heterogeneous printed probe radial density that influences hybridization spatially and temporally via radial hemispherical diffusion of dye-labeled target from the outside edge of the spot to the interior. FRET of the surface-captured target occurs irrespective of the probe/target fluorophore position, resulting from excess printed probe density and spot thickness. Both heterogeneous probe density distributions in printed spots and the fluorophore position on short DNA oligomers influence duplex formation kinetics, hybridization efficiencies, and overall fluorescence intensity end points in surface-capture formats. This analysis is important to understanding, controlling, and quantifying the array assay signal essential to reliable application of the surface-capture format.


Asunto(s)
ADN/análisis , ADN/química , Microscopía Fluorescente/métodos , Hibridación de Ácido Nucleico , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Carbocianinas/química , Sondas de ADN/química , Colorantes Fluorescentes/química , Cinética , Microscopía Confocal , Fotoblanqueo , Factores de Tiempo
4.
FASEB J ; 26(4): 1442-51, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22202674

RESUMEN

To investigate the effects of the predominant nonprotein thiol, glutathione (GSH), on redox homeostasis, we employed complementary pharmacological and genetic strategies to determine the consequences of both loss- and gain-of-function GSH content in vitro. We monitored the redox events in the cytosol and mitochondria using reduction-oxidation sensitive green fluorescent protein (roGFP) probes and the level of reduced/oxidized thioredoxins (Trxs). Either H(2)O(2) or the Trx reductase inhibitor 1-chloro-2,4-dinitrobenzene (DNCB), in embryonic rat heart (H9c2) cells, evoked 8 or 50 mV more oxidizing glutathione redox potential, E(hc) (GSSG/2GSH), respectively. In contrast, N-acetyl-L-cysteine (NAC) treatment in H9c2 cells, or overexpression of either the glutamate cysteine ligase (GCL) catalytic subunit (GCLC) or GCL modifier subunit (GCLM) in human embryonic kidney 293 T (HEK293T) cells, led to 3- to 4-fold increase of GSH and caused 7 or 12 mV more reducing E(hc), respectively. This condition paradoxically increased the level of mitochondrial oxidation, as demonstrated by redox shifts in mitochondrial roGFP and Trx2. Lastly, either NAC treatment (EC(50) 4 mM) or either GCLC or GCLM overexpression exhibited increased cytotoxicity and the susceptibility to the more reducing milieu was achieved at decreased levels of ROS. Taken together, our findings reveal a novel mechanism by which GSH-dependent reductive stress triggers mitochondrial oxidation and cytotoxicity.


Asunto(s)
Glutatión/metabolismo , Mitocondrias/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Acetilcisteína/farmacología , Animales , Citosol/metabolismo , Dinitroclorobenceno/farmacología , Depuradores de Radicales Libres/farmacología , Glutamato-Cisteína Ligasa/metabolismo , Disulfuro de Glutatión/metabolismo , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Corazón/efectos de los fármacos , Corazón/embriología , Homeostasis , Humanos , Peróxido de Hidrógeno/farmacología , Indicadores y Reactivos/farmacología , Miocardio/citología , Miocardio/metabolismo , Oxidantes/farmacología , Subunidades de Proteína/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Tiorredoxinas/metabolismo
5.
Cancer Immunol Immunother ; 61(7): 1113-24, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22198309

RESUMEN

Cancer survivors often relapse due to evolving drug-resistant clones and repopulating tumor stem cells. Our preclinical study demonstrated that terminal cancer patient's lymphocytes can be converted from tolerant bystanders in vivo into effective cytotoxic T-lymphocytes in vitro killing patient's own tumor cells containing drug-resistant clones and tumor stem cells. We designed a clinical trial combining peginterferon α-2b with imatinib for treatment of stage III/IV gastrointestinal stromal tumor (GIST) with the rational that peginterferon α-2b serves as danger signals to promote antitumor immunity while imatinib's effective tumor killing undermines tumor-induced tolerance and supply tumor-specific antigens in vivo without leukopenia, thus allowing for proper dendritic cell and cytotoxic T-lymphocyte differentiation toward Th1 response. Interim analysis of eight patients demonstrated significant induction of IFN-γ-producing-CD8(+), -CD4(+), -NK cell, and IFN-γ-producing-tumor-infiltrating-lymphocytes, signifying significant Th1 response and NK cell activation. After a median follow-up of 3.6 years, complete response (CR) + partial response (PR) = 100%, overall survival = 100%, one patient died of unrelated illness while in remission, six of seven evaluable patients are either in continuing PR/CR (5 patients) or have progression-free survival (PFS, 1 patient) exceeding the upper limit of the 95% confidence level of the genotype-specific-PFS of the phase III imatinib-monotherapy (CALGB150105/SWOGS0033), demonstrating highly promising clinical outcomes. The current trial is closed in preparation for a larger future trial. We conclude that combination of targeted therapy and immunotherapy is safe and induced significant Th1 response and NK cell activation and demonstrated highly promising clinical efficacy in GIST, thus warranting development in other tumor types.


Asunto(s)
Neoplasias Gastrointestinales/terapia , Tumores del Estroma Gastrointestinal/terapia , Interferón-alfa/administración & dosificación , Piperazinas/administración & dosificación , Polietilenglicoles/administración & dosificación , Pirimidinas/administración & dosificación , Anciano , Anciano de 80 o más Años , Benzamidas , Supervivencia sin Enfermedad , Neoplasias Gastrointestinales/inmunología , Tumores del Estroma Gastrointestinal/inmunología , Humanos , Mesilato de Imatinib , Inmunoterapia/métodos , Interferón alfa-2 , Interferón-alfa/inmunología , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Linfocitos/inmunología , Persona de Mediana Edad , Proteínas Recombinantes/administración & dosificación , Recurrencia , Linfocitos T Citotóxicos/inmunología
6.
Proc Natl Acad Sci U S A ; 107(29): 12889-94, 2010 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-20616062

RESUMEN

The ESCRT pathway helps mediate the final abscission step of cytokinesis in mammals and archaea. In mammals, two early acting proteins of the ESCRT pathway, ALIX and TSG101, are recruited to the midbody through direct interactions with the phosphoprotein CEP55. CEP55 resides at the centrosome through most of the cell cycle but then migrates to the midbody at the start of cytokinesis, suggesting that the ESCRT pathway may also have centrosomal links. Here, we have systematically analyzed the requirements for late-acting mammalian ESCRT-III and VPS4 proteins at different stages of mitosis and cell division. We found that depletion of VPS4A, VPS4B, or any of the 11 different human ESCRT-III (CHMP) proteins inhibited abscission. Remarkably, depletion of individual ESCRT-III and VPS4 proteins also altered centrosome and spindle pole numbers, producing multipolar spindles (most ESCRT-III/VPS4 proteins) or monopolar spindles (CHMP2A or CHMP5) and causing defects in chromosome segregation and nuclear morphology. VPS4 proteins concentrated at spindle poles during mitosis and then at midbodies during cytokinesis, implying that these proteins function directly at both sites. We conclude that ESCRT-III/VPS4 proteins function at centrosomes to help regulate their maintenance or proliferation and then at midbodies during abscission, thereby helping ensure the ordered progression through the different stages of cell division.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Centrosoma/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Huso Acromático/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfatasas/deficiencia , Biomarcadores de Tumor/metabolismo , Supervivencia Celular , Citocinesis , ADN/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/deficiencia , Células HeLa , Humanos , Imagenología Tridimensional , Mitosis , Transporte de Proteínas , Factores de Tiempo , ATPasas de Translocación de Protón Vacuolares
7.
J Vis Exp ; (40)2010 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-20548280

RESUMEN

Changes in cellular organization and chromosome dynamics that occur during mitosis are tightly coordinated to ensure accurate inheritance of genomic and cellular content. Hallmark events of mitosis, such as chromosome movement, can be readily tracked on an individual cell basis using time-lapse fluorescence microscopy of mammalian cell lines expressing specific GFP-tagged proteins. In combination with RNAi-based depletion, this can be a powerful method for pinpointing the stage(s) of mitosis where defects occur after levels of a particular protein have been lowered. In this protocol, we present a basic method for assessing the effect of depleting a potential mitotic regulatory protein on the timing of mitosis. Cells are transfected with siRNA, placed in a stage-top incubation chamber, and imaged using an automated fluorescence microscope. We describe how to use software to set up a time-lapse experiment, how to process the image sequences to make either still-image montages or movies, and how to quantify and analyze the timing of mitotic stages using a cell-line expressing mCherry-tagged histone H2B. Finally, we discuss important considerations for designing a time-lapse experiment. This strategy is complementary to other approaches and offers the advantages of 1) sensitivity to changes in kinetics that might not be observed when looking at cells as a population and 2) analysis of mitosis without the need to synchronize the cell cycle using drug treatments. The visual information from such imaging experiments not only allows the sub-stages of mitosis to be assessed, but can also provide unexpected insight that would not be apparent from cell cycle analysis by FACS.


Asunto(s)
Procesamiento de Imagen Asistido por Computador/métodos , Microscopía Fluorescente/métodos , Mitosis/genética , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Transfección/métodos , Histonas/genética , Humanos
8.
Toxicol Appl Pharmacol ; 236(1): 71-7, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19371621

RESUMEN

AIMS: Our goal was to determine if clinically relevant concentrations of aqueous extract of cigarette smoke (CSE) have direct deleterious effects on ventricular myocytes during simulated ischemia, and to investigate the mechanisms involved. METHODS: CSE was prepared with a smoking chamber. Ischemia was simulated by metabolic inhibition (MI) with cyanide (CN) and 0 glucose. Adult rabbit and mouse ventricular myocyte [Ca(2+)](i) was measured by flow cytometry using fluo-3. Mitochondrial [Ca(2+)] was measured with confocal microscopy, and Rhod-2 fluorescence. The mitochondrial permeability transition (MPT) was detected by TMRM fluorescence and myocyte contracture. Myocyte oxidative stress was quantified by dichlorofluorescein (DCF) fluorescence with confocal microscopy. RESULTS: CSE 0.1% increased myocyte contracture caused by MI. The nicotine concentration (HPLC) in 0.1% CSE was 15 ng/ml, similar to that in humans after smoking cigarettes. CSE 0.1% increased mitochondrial Ca(2+) uptake, and increased the susceptibility of mitochondria to the MPT. CSE 0.1% increased DCF fluorescence in isolated myocytes, and increased [Ca(2+)](i) in paced myocytes exposed to 2.0 mM CN, 0 glucose (P-MI). These effects were inhibited by the superoxide scavenger Tiron. The effect of CSE on [Ca(2+)](i) during P-MI was also prevented by ranolazine. CONCLUSIONS: CSE in clinically relevant concentrations increases myocyte [Ca(2+)](i) during simulated ischemia, and increases myocyte susceptibility to the MPT. These effects appear to be mediated at least in part by oxidative radicals in CSE, and likely contribute to the effects of cigarette smoke to increase myocardial infarct size, and to decrease angina threshold.


Asunto(s)
Contracción Miocárdica/efectos de los fármacos , Isquemia Miocárdica/complicaciones , Miocitos Cardíacos/efectos de los fármacos , Humo/efectos adversos , Fumar/efectos adversos , Sal Disódica del Ácido 1,2-Dihidroxibenceno-3,5-Disulfónico/farmacología , Acetanilidas/farmacología , Angina de Pecho/etiología , Angina de Pecho/metabolismo , Animales , Calcio/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Depuradores de Radicales Libres/farmacología , Glucosa/metabolismo , Ventrículos Cardíacos/efectos de los fármacos , Ratones , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/efectos de los fármacos , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Infarto del Miocardio/etiología , Infarto del Miocardio/fisiopatología , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/fisiopatología , Miocitos Cardíacos/metabolismo , Nicotina/análisis , Piperazinas/farmacología , Conejos , Ranolazina , Especies Reactivas de Oxígeno/metabolismo , Humo/análisis , Factores de Tiempo
9.
Blood ; 113(25): 6419-27, 2009 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-19221037

RESUMEN

Neutrophils are highly specialized innate effector cells that have evolved for killing of pathogens. Human neonates have a common multifactorial syndrome of neutrophil dysfunction that is incompletely characterized and contributes to sepsis and other severe infectious complications. We identified a novel defect in the antibacterial defenses of neonates: inability to form neutrophil extracellular traps (NETs). NETs are lattices of extracellular DNA, chromatin, and antibacterial proteins that mediate extracellular killing of microorganisms and are thought to form via a unique death pathway signaled by nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-generated reactive oxygen species (ROS). We found that neutrophils from term and preterm infants fail to form NETs when activated by inflammatory agonists-in contrast to leukocytes from healthy adults. The deficiency in NET formation is paralleled by a previously unrecognized deficit in extracellular bacterial killing. Generation of ROSs did not complement the defect in NET formation by neonatal neutrophils, as it did in adult cells with inactivated NADPH oxidase, demonstrating that ROSs are necessary but not sufficient signaling intermediaries and identifying a deficiency in linked or downstream pathways in neonatal leukocytes. Impaired NET formation may be a critical facet of a common developmental immunodeficiency that predisposes newborn infants to infection.


Asunto(s)
Actividad Bactericida de la Sangre , Recién Nacido/inmunología , Recien Nacido Prematuro/inmunología , Sustancias Macromoleculares/inmunología , Neutrófilos/patología , Adulto , Envejecimiento/inmunología , Cromatina/fisiología , ADN/fisiología , Susceptibilidad a Enfermedades , Espacio Extracelular , Sangre Fetal/citología , Sangre Fetal/inmunología , Humanos , Infecciones/inmunología , Lipopolisacáridos/farmacología , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Factor de Activación Plaquetaria/farmacología , Glicoproteínas de Membrana Plaquetaria/biosíntesis , Glicoproteínas de Membrana Plaquetaria/genética , ARN Mensajero/biosíntesis , Receptores Acoplados a Proteínas G/biosíntesis , Receptores Acoplados a Proteínas G/genética , Estallido Respiratorio , Receptor Toll-Like 4/biosíntesis , Receptor Toll-Like 4/genética
10.
EMBO J ; 26(19): 4215-27, 2007 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-17853893

RESUMEN

TSG101 and ALIX both function in HIV budding and in vesicle formation at the multivesicular body (MVB), where they interact with other Endosomal Sorting Complex Required for Transport (ESCRT) pathway factors required for release of viruses and vesicles. Proteomic analyses revealed that ALIX and TSG101/ESCRT-I also bind a series of proteins involved in cytokinesis, including CEP55, CD2AP, ROCK1, and IQGAP1. ALIX and TSG101 concentrate at centrosomes and are then recruited to the midbodies of dividing cells through direct interactions between the central CEP55 'hinge' region and GPP-based motifs within TSG101 and ALIX. ESCRT-III and VPS4 proteins are also recruited, indicating that much of the ESCRT pathway localizes to the midbody. Depletion of ALIX and TSG101/ESCRT-I inhibits the abscission step of HeLa cell cytokinesis, as does VPS4 overexpression, confirming a requirement for these proteins in cell division. Furthermore, ALIX point mutants that block CEP55 and CHMP4/ESCRT-III binding also inhibit abscission, indicating that both interactions are essential. These experiments suggest that the ESCRT pathway may be recruited to facilitate analogous membrane fission events during HIV budding, MVB vesicle formation, and the abscission stage of cytokinesis.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Citocinesis/fisiología , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/metabolismo , Secuencias de Aminoácidos/fisiología , Animales , Células COS , Proteínas de Unión al Calcio/genética , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/genética , Centrosoma/metabolismo , Chlorocebus aethiops , Proteínas de Unión al ADN/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte , Endosomas/genética , Endosomas/metabolismo , VIH/fisiología , Células HeLa , Humanos , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Unión Proteica/fisiología , Estructura Terciaria de Proteína/fisiología , Factores de Transcripción/genética , Ensamble de Virus/fisiología , Internalización del Virus
11.
J Neurosci ; 25(12): 3199-208, 2005 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-15788777

RESUMEN

A Drosophila forward genetic screen for mutants with defective synaptic development identified bad reception (brec). Homozygous brec mutants are embryonic lethal, paralyzed, and show no detectable synaptic transmission at the glutamatergic neuromuscular junction (NMJ). Genetic mapping, complementation tests, and genomic sequencing show that brec mutations disrupt a previously uncharacterized ionotropic glutamate receptor subunit, named here "GluRIID." GluRIID is expressed in the postsynaptic domain of the NMJ, as well as widely throughout the synaptic neuropil of the CNS. In the NMJ of null brec mutants, all known glutamate receptor subunits are undetectable by immunocytochemistry, and all functional glutamate receptors are eliminated. Thus, we conclude that GluRIID is essential for the assembly and/or stabilization of glutamate receptors in the NMJ. In null brec mutant embryos, the frequency of periodic excitatory currents in motor neurons is significantly reduced, demonstrating that CNS motor pattern activity is regulated by GluRIID. Although synaptic development and molecular differentiation appear otherwise unperturbed in null mutants, viable hypomorphic brec mutants display dramatically undergrown NMJs by the end of larval development, suggesting that GluRIID-dependent central pattern activity regulates peripheral synaptic growth. These studies reveal GluRIID as a newly identified glutamate receptor subunit that is essential for glutamate receptor assembly/stabilization in the peripheral NMJ and required for properly patterned motor output in the CNS.


Asunto(s)
Proteínas de Drosophila/fisiología , Unión Neuromuscular/fisiología , Neurópilo/metabolismo , Receptores de Glutamato/fisiología , Animales , Animales Modificados Genéticamente , Drosophila , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/metabolismo , Estimulación Eléctrica/métodos , Embrión no Mamífero , Potenciales Postsinápticos Excitadores/genética , Potenciales Postsinápticos Excitadores/efectos de la radiación , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica/métodos , Microscopía Confocal/métodos , Biología Molecular/métodos , Técnicas de Placa-Clamp/métodos , Subunidades de Proteína/deficiencia , Subunidades de Proteína/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptores de Glutamato/deficiencia , Proteínas Supresoras de Tumor/metabolismo , Proteína Letal Asociada a bcl/deficiencia
12.
Dev Dyn ; 229(3): 651-7, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14991720

RESUMEN

Multi-color whole-mount in situ hybridization is a powerful technique for comparing the spatial expression patterns of two or more genes in developing embryos. We have developed an amplified triple-label whole-mount fluorescence in situ hybridization (FISH) protocol that permits detection of three different mRNAs in a single embryo. Our protocol uses simultaneous in situ hybridization to haptenylated riboprobes, followed by sequential antibody detection using anti-hapten antibodies conjugated to horseradish peroxidase, and the tyramide signal amplification (TSA) fluorescence detection system. Conventional fluorescence microscopy identifies areas of overlapping gene expression at the tissue level, whereas confocal fluorescence microscopy permits single-cell resolution and differentiates specialized cell types within a given tissue. This protocol will provide researchers engaged in the use of FISH with a solid starting point for adapting their own in situ hybridization protocols, either alone or in combination with immunohistochemistry or green fluorescence protein colocalization.


Asunto(s)
Biología Evolutiva/métodos , Regulación del Desarrollo de la Expresión Génica , Hibridación Fluorescente in Situ/métodos , Animales , Embrión de Pollo , Proteínas de Unión al ADN/metabolismo , Glicoproteínas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Mesodermo/metabolismo , Microscopía Confocal , Microscopía Fluorescente/métodos , Factor de Transcripción PAX2 , Factores de Tiempo , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA