Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neurochem ; 128(5): 686-700, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24117625

RESUMEN

The WWC1 gene has been genetically associated with human episodic memory performance, and its product KIdney/BRAin protein (KIBRA) has been shown to interact with the atypical protein kinase protein kinase M ζ (PKMζ). Although recently challenged, PKMζ remains a candidate postsynaptic regulator of memory maintenance. Here, we show that PKMζ is subject to rapid proteasomal degradation and that KIBRA is both necessary and sufficient to counteract this process, thus stabilizing the kinase and maintaining its function for a prolonged time. We define the binding sequence on KIBRA, a short amino acid motif near the C-terminus. Both hippocampal knock-down of KIBRA in rats and KIBRA knock-out in mice result in decreased learning and memory performance in spatial memory tasks supporting the notion that KIBRA is a player in episodic memory. Interestingly, decreased memory performance is accompanied by decreased PKMζ protein levels. We speculate that the stabilization of synaptic PKMζ protein levels by KIBRA may be one mechanism by which KIBRA acts in memory maintenance. KIBRA/WWC1 has been genetically associated with human episodic memory. KIBRA has been shown to be post-synaptically localized, but its function remained obscure. Here, we show that KIBRA shields PKMζ, a kinase previously linked to memory maintenance, from proteasomal degradation via direct interaction. KIBRA levels in the rodent hippocampus correlate closely both to spatial memory performance in rodents and to PKMζ levels. Our findings support a role for KIBRA in memory, and unveil a novel function for this protein.


Asunto(s)
Proteínas Portadoras/fisiología , Proteínas Co-Represoras/fisiología , Aprendizaje/fisiología , Memoria/fisiología , Proteína Quinasa C/fisiología , Secuencia de Aminoácidos , Animales , Reacción de Prevención/fisiología , Conducta Animal/fisiología , Western Blotting , Proteínas Portadoras/metabolismo , Proteínas Co-Represoras/metabolismo , Dependovirus/genética , Prueba de Complementación Genética , Hipocampo/metabolismo , Hipocampo/fisiología , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Datos de Secuencia Molecular , Fosfoproteínas , Reacción en Cadena de la Polimerasa , Unión Proteica , Proteína Quinasa C/metabolismo , Ratas , Ratas Wistar , Técnicas Estereotáxicas
2.
J Neurochem ; 113(4): 930-42, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20202082

RESUMEN

Granulocyte-colony stimulating factor (G-CSF) is a potent hematopoietic factor that drives differentiation of neutrophilic granulocytes. We have recently shown that G-CSF also acts as a neuronal growth factor, protects neurons in vitro and in vivo, and has regenerative potential in various neurological disease models. Spinal cord injury (SCI) following trauma or secondary to skeletal instability is a terrible condition with no effective therapies available at present. In this study, we show that the G-CSF receptor is up-regulated upon experimental SCI and that G-CSF improves functional outcome in a partial dissection model of SCI. G-CSF significantly decreases apoptosis in an experimental partial spinal transsection model in the mouse and increases expression of the anti-apoptotic G-CSF target gene Bcl-X(L). In vitro, G-CSF enhances neurite outgrowth and branching capacity of hippocampal neurons. In vivo, G-CSF treatment results in improved functional connectivity of the injured spinal cord as measured by Mn(2+)-enhanced MRI. G-CSF also increased length of the dorsal corticospinal tract and density of serotonergic fibers cranial to the lesion center. Mice treated systemically with G-CSF as well as transgenic mice over-expressing G-CSF in the CNS exhibit a strong improvement in functional outcome as measured by the BBB score and gridwalk analysis. We show that G-CSF improves outcome after experimental SCI by counteracting apoptosis, and enhancing connectivity in the injured spinal cord. We conclude that G-CSF constitutes a promising and feasible new therapy option for SCI.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/farmacología , Factores de Crecimiento Nervioso/farmacología , Fármacos Neuroprotectores/farmacología , Traumatismos de la Médula Espinal/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Conos de Crecimiento/efectos de los fármacos , Conos de Crecimiento/fisiología , Conos de Crecimiento/ultraestructura , Ratones , Factores de Crecimiento Nervioso/uso terapéutico , Neuritas/efectos de los fármacos , Neuritas/ultraestructura , Fármacos Neuroprotectores/uso terapéutico , Tractos Piramidales/efectos de los fármacos , Tractos Piramidales/lesiones , Tractos Piramidales/fisiología , Ratas , Ratas Wistar , Receptores de Factor Estimulante de Colonias de Granulocito/efectos de los fármacos , Receptores de Factor Estimulante de Colonias de Granulocito/metabolismo , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/fisiopatología , Resultado del Tratamiento , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Proteína bcl-X/efectos de los fármacos , Proteína bcl-X/metabolismo
3.
Brain ; 131(Pt 12): 3335-47, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18835867

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease that results in progressive loss of motoneurons, motor weakness and death within 1-5 years after disease onset. Therapeutic options remain limited despite a substantial number of approaches that have been tested clinically. In particular, various neurotrophic factors have been investigated. Failure in these trials has been largely ascribed to problems of insufficient dosing or inability to cross the blood-brain barrier (BBB). We have recently uncovered the neurotrophic properties of the haematopoietic protein granulocyte-colony stimulating factor (G-CSF). The protein is clinically well tolerated and crosses the intact BBB. This study examined the potential role of G-CSF in motoneuron diseases. We investigated the expression of the G-CSF receptor in motoneurons and studied effects of G-CSF in a motoneuron cell line and in the SOD1(G93A) transgenic mouse model. The neurotrophic growth factor was applied both by continuous subcutaneous delivery and CNS-targeted transgenic overexpression. This study shows that given at the stage of the disease where muscle denervation is already evident, G-CSF leads to significant improvement in motor performance, delays the onset of severe motor impairment and prolongs overall survival of SOD1(G93A)tg mice. The G-CSF receptor is expressed by motoneurons and G-CSF protects cultured motoneuronal cells from apoptosis. In ALS mice, G-CSF increased survival of motoneurons and decreased muscular denervation atrophy. We conclude that G-CSF is a novel neurotrophic factor for motoneurons that is an attractive and feasible drug candidate for the treatment of ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Femenino , Filgrastim , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Factor Estimulante de Colonias de Granulocitos/farmacología , Humanos , Infusiones Subcutáneas , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/metabolismo , Mutación , Receptores de Factor Estimulante de Colonias de Granulocito/metabolismo , Proteínas Recombinantes , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Médula Espinal/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa-1 , Resultado del Tratamiento
4.
BMC Dev Biol ; 8: 32, 2008 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-18371196

RESUMEN

BACKGROUND: Granulocyte colony-stimulating (G-CSF) factor is a well-known hematopoietic growth factor stimulating the proliferation and differentiation of myeloid progenitors. Recently, we uncovered that G-CSF acts also as a neuronal growth factor in the brain, which promotes adult neural precursor differentiation and enhances regeneration of the brain after insults. In adults, the receptor for G-CSF is predominantly expressed in neurons in many brain areas. We also described expression in neurogenic regions of the adult brain, such as the subventricular zone and the subgranular layer of the dentate gyrus. In addition, we found close co-localization of the G-CSF receptor and its ligand G-CSF. Here we have conducted a systematic expression analysis of G-CSF receptor and its ligand in the developing embryo. RESULTS: Outside the central nervous system (CNS) we found G-CSF receptor expression in blood vessels, muscles and their respective precursors and neurons. The expression of the G-CSF receptor in the developing CNS was most prominent in radial glia cells. CONCLUSION: Our data imply that in addition to the function of G-CSF and its receptor in adult neurogenesis, this system also has a role in embryonic neurogenesis and nervous system development.


Asunto(s)
Encéfalo/embriología , Regulación del Desarrollo de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos/genética , Neuroglía/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocito/genética , Animales , Embrión de Mamíferos , Factor Estimulante de Colonias de Granulocitos/metabolismo , Inmunohistoquímica , Proteínas de Filamentos Intermediarios/genética , Proteínas de Filamentos Intermediarios/metabolismo , Morfogénesis/genética , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Nestina , ARN Mensajero/biosíntesis , Ratas , Ratas Wistar , Receptores de Factor Estimulante de Colonias de Granulocito/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
5.
J Cereb Blood Flow Metab ; 28(1): 29-43, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17457367

RESUMEN

Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a hematopoietic cytokine responsible for the proliferation, differentiation, and maturation of cells of the myeloid lineage, which was cloned more than 20 years ago. Here we uncovered a novel function of GM-CSF in the central nervous system (CNS). We identified the GM-CSF alpha-receptor as an upregulated gene in a screen for ischemia-induced genes in the cortex. This receptor is broadly expressed on neurons throughout the brain together with its ligand and induced by ischemic insults. In primary cortical neurons and human neuroblastoma cells, GM-CSF counteracts programmed cell death and induces BCL-2 and BCL-Xl expression in a dose- and time-dependent manner. Of the signaling pathways studied, GM-CSF most prominently induced the PI3K-Akt pathway, and inhibition of Akt strongly decreased antiapoptotic activity. Intravenously given GM-CSF passes the blood-brain barrier, and decreases infarct damage in two different experimental stroke models (middle cerebral artery occlusion (MCAO), and combined common carotid/distal MCA occlusion) concomitant with induction of BCL-Xl expression. Thus, GM-CSF acts as a neuroprotective protein in the CNS. This finding is remarkably reminiscent of the recently discovered functionality of two other hematopoietic factors, erythropoietin and granulocyte colony-stimulating factor in the CNS. The identification of a third hematopoietic factor acting as a neurotrophic factor in the CNS suggests a common principle in the functional evolution of these factors. Clinically, GM-CSF now broadens the repertoire of hematopoietic factors available as novel drug candidates for stroke and neurodegenerative diseases.


Asunto(s)
Apoptosis/efectos de los fármacos , Infarto Encefálico/tratamiento farmacológico , Isquemia Encefálica/tratamiento farmacológico , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Fármacos Neuroprotectores/farmacología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Animales , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Infarto Encefálico/metabolismo , Infarto Encefálico/patología , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Humanos , Masculino , Células Mieloides/metabolismo , Células Mieloides/patología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neuronas/metabolismo , Neuronas/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Long-Evans , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Factores de Tiempo , Regulación hacia Arriba/efectos de los fármacos , Proteína bcl-X/biosíntesis
6.
J Clin Invest ; 115(8): 2083-98, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16007267

RESUMEN

G-CSF is a potent hematopoietic factor that enhances survival and drives differentiation of myeloid lineage cells, resulting in the generation of neutrophilic granulocytes. Here, we show that G-CSF passes the intact blood-brain barrier and reduces infarct volume in 2 different rat models of acute stroke. G-CSF displays strong anti-apoptotic activity in mature neurons and activates multiple cell survival pathways. Both G-CSF and its receptor are widely expressed by neurons in the CNS, and their expression is induced by ischemia, which suggests an autocrine protective signaling mechanism. Surprisingly, the G-CSF receptor was also expressed by adult neural stem cells, and G-CSF induced neuronal differentiation in vitro. G-CSF markedly improved long-term behavioral outcome after cortical ischemia, while stimulating neural progenitor response in vivo, providing a link to functional recovery. Thus, G-CSF is an endogenous ligand in the CNS that has a dual activity beneficial both in counteracting acute neuronal degeneration and contributing to long-term plasticity after cerebral ischemia. We therefore propose G-CSF as a potential new drug for stroke and neurodegenerative diseases.


Asunto(s)
Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Receptores de Factor Estimulante de Colonias de Granulocito/biosíntesis , Células Madre/metabolismo , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Barrera Hematoencefálica/metabolismo , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Factor Estimulante de Colonias de Granulocitos/biosíntesis , Células Precursoras de Granulocitos/metabolismo , Humanos , Ligandos , Masculino , Tejido Nervioso/metabolismo , Tejido Nervioso/patología , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Neutrófilos/metabolismo , Ratas , Ratas Wistar , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología
7.
Neurosci Lett ; 430(2): 138-41, 2008 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-18035491

RESUMEN

Cerebral ischemia triggers inflammation and apoptosis, and the transcription factor NF-kappaB is a key regulator of both events. Here, we report on the induction of the peptidoglycan recognition protein-S (PGRP-S) in a mouse model of cerebral ischemia. Upregulation was reduced if the NF-kappaB subunit RelA was conditionally deleted in the brain. Regulation of PGRP-S transcription by RelA was confirmed in vitro. Cotransfection of a RelA expression plasmid stimulated the expression of a PGRP-S luciferase fusion gene. Mutation of two NF-kappaB response elements in the PGRP-S promoter disrupted stimulation by RelA. To investigate the function of PGRP-S in cerebral ischemia, we subjected PGRP-S(-/-) mice to cerebral ischemia. However, there was no difference in the infarct size in PGRP-S-deficient mice compared to controls. In summary, the data show that PGRP-S is induced in cerebral ischemia by RelA, but its role in ischemia is unclear.


Asunto(s)
Proteínas Portadoras/metabolismo , Factor de Transcripción ReIA/fisiología , Regulación hacia Arriba/fisiología , Análisis de Varianza , Animales , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Proteínas Portadoras/genética , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Mutación , Células PC12 , Ratas , Transfección , Regulación hacia Arriba/efectos de los fármacos
8.
BMC Genomics ; 8: 370, 2007 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17937787

RESUMEN

BACKGROUND: The different physiological repertoire of CA3 and CA1 neurons in the hippocampus, as well as their differing behaviour after noxious stimuli are ultimately based upon differences in the expressed genome. We have compared CA3 and CA1 gene expression in the uninjured brain, and after cerebral ischemia using laser microdissection (LMD), RNA amplification, and array hybridization. RESULTS: Profiling in CA1 vs. CA3 under normoxic conditions detected more than 1000 differentially expressed genes that belong to different, physiologically relevant gene ontology groups in both cell types. The comparison of each region under normoxic and ischemic conditions revealed more than 5000 ischemia-regulated genes for each individual cell type. Surprisingly, there was a high co-regulation in both regions. In the ischemic state, only about 100 genes were found to be differentially expressed in CA3 and CA1. The majority of these genes were also different in the native state. A minority of interesting genes (e.g. inhibinbetaA) displayed divergent expression preference under native and ischemic conditions with partially opposing directions of regulation in both cell types. CONCLUSION: The differences found in two morphologically very similar cell types situated next to each other in the CNS are large providing a rational basis for physiological differences. Unexpectedly, the genomic response to ischemia is highly similar in these two neuron types, leading to a substantial attenuation of functional genomic differences in these two cell types. Also, the majority of changes that exist in the ischemic state are not generated de novo by the ischemic stimulus, but are preexistant from the genomic repertoire in the native situation. This unexpected influence of a strong noxious stimulus on cell-specific gene expression differences can be explained by the activation of a cell-type independent conserved gene-expression program. Our data generate both novel insights into the relation of the quiescent and stimulus-induced transcriptome in different cells, and provide a large dataset to the research community, both for mapping purposes, as well as for physiological and pathophysiological research.


Asunto(s)
Isquemia Encefálica/genética , Genoma , Neuronas/metabolismo , Animales , Perfilación de la Expresión Génica , Inmunohistoquímica , Ratas
9.
BMC Neurosci ; 8: 88, 2007 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-17953750

RESUMEN

BACKGROUND: Granulocyte-macrophage colony stimulating factor (GM-CSF) is a hematopoietic growth factor involved in the generation of granulocytes, macrophages, and dendritic cells from hematopoietic progenitor cells. We have recently demonstrated that GM-CSF has anti-apoptotic functions on neurons, and is neuroprotective in animal stroke models. RESULTS: The GM-CSF receptor alpha is expressed on adult neural stem cells in the rodent brain, and in culture. Addition of GM-CSF to NSCs in vitro increased neuronal differentiation in a dose-dependent manner as determined by quantitative PCR, reporter gene assays, and FACS analysis. CONCLUSION: Similar to the hematopoietic factor Granulocyte-colony stimulating factor (G-CSF), GM-CSF stimulates neuronal differentiation of adult NSCs. These data highlight the astonishingly similar functions of major hematopoietic factors in the brain, and raise the clinical attractiveness of GM-CSF as a novel drug for neurological disorders.


Asunto(s)
Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Diferenciación Celular/fisiología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Neuronas/citología , Neuronas/metabolismo , Animales , Células Cultivadas , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Masculino , Ratas , Ratas Wistar
10.
BMC Biol ; 4: 36, 2006 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-17049076

RESUMEN

BACKGROUND: Granulocyte-colony stimulating factor (G-CSF) is known as a powerful regulator of white blood cell proliferation and differentiation in mammals. We, and others, have shown that G-CSF is effective in treating cerebral ischemia in rodents, both relating to infarct size as well as functional recovery. G-CSF and its receptor are expressed by neurons, and G-CSF regulates apoptosis and neurogenesis, providing a rational basis for its beneficial short- and long-term actions in ischemia. In addition, G-CSF may contribute to re-endothelialisation and arteriogenesis in the vasculature of the ischemic penumbra. In addition to these trophic effects, G-CSF is a potent neuroprotective factor reliably reducing infarct size in different stroke models. RESULTS: Here, we have further delayed treatment and studied effects of G-CSF on infarct volume in the middle cerebral artery occlusion (MCAO) model and functional outcome in the cortical photothrombotic model. In the MCAO model, we applied a single dose of 60 microg/kg bodyweight G-CSF in rats 4 h after onset of ischemia. Infarct volume was determined 24 h after onset of ischemia. In the rat photothrombotic model, we applied 10 microg/kg bodyweight G-CSF daily for a period of 10 days starting either 24 or 72 h after induction of ischemia. G-CSF both decreased acute infarct volume in the MCAO model, and improved recovery in the photothrombotic model at delayed timepoints. CONCLUSION: These data further strengthen G-CSF's profile as a unique candidate stroke drug, and provide an experimental basis for application of G-CSF in the post-stroke recovery phase.


Asunto(s)
Infarto Cerebral/tratamiento farmacológico , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Fármacos Neuroprotectores/administración & dosificación , Fármacos Neuroprotectores/uso terapéutico , Animales , Esquema de Medicación , Masculino , Equilibrio Postural/efectos de los fármacos , Desempeño Psicomotor/efectos de los fármacos , Ratas , Ratas Wistar
11.
Nat Commun ; 7: 13296, 2016 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-27897270

RESUMEN

Sequencing of a single-cell genome requires DNA amplification, a process prone to introducing bias and errors into the amplified genome. Here we introduce a novel multiple displacement amplification (MDA) method based on the unique DNA primase features of Thermus thermophilus (Tth) PrimPol. TthPrimPol displays a potent primase activity preferring dNTPs as substrates unlike conventional primases. A combination of TthPrimPol's unique ability to synthesize DNA primers with the highly processive Phi29 DNA polymerase (Φ29DNApol) enables near-complete whole genome amplification from single cells. This novel method demonstrates superior breadth and evenness of genome coverage, high reproducibility, excellent single-nucleotide variant (SNV) detection rates with low allelic dropout (ADO) and low chimera formation as exemplified by sequencing HEK293 cells. Moreover, copy number variant (CNV) calling yields superior results compared with random primer-based MDA methods. The advantages of this method, which we named TruePrime, promise to facilitate and improve single-cell genomic analysis.


Asunto(s)
Genoma Humano , Reacción en Cadena de la Polimerasa/métodos , Análisis de la Célula Individual , Alelos , Secuencia de Aminoácidos , Secuencia de Bases , ADN/genética , Variaciones en el Número de Copia de ADN/genética , ADN Primasa/química , ADN Primasa/metabolismo , ADN Polimerasa Dirigida por ADN/metabolismo , Humanos , Reproducibilidad de los Resultados , Análisis de Secuencia de ADN , Moldes Genéticos , Thermus thermophilus/enzimología
12.
Cancer Cell ; 28(5): 610-622, 2015 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-26481148

RESUMEN

While recombinant human erythropoietin (rhEpo) has been widely used to treat anemia in cancer patients, concerns about its adverse effects on patient survival have emerged. A lack of correlation between expression of the canonical EpoR and rhEpo's effects on cancer cells prompted us to consider the existence of an alternative Epo receptor. Here, we identified EphB4 as an Epo receptor that triggers downstream signaling via STAT3 and promotes rhEpo-induced tumor growth and progression. In human ovarian and breast cancer samples, expression of EphB4 rather than the canonical EpoR correlated with decreased disease-specific survival in rhEpo-treated patients. These results identify EphB4 as a critical mediator of erythropoietin-induced tumor progression and further provide clinically significant dimension to the biology of erythropoietin.


Asunto(s)
Neoplasias de la Mama/genética , Eritropoyetina/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Ováricas/genética , Receptor EphB4/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , Western Blotting , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Progresión de la Enfermedad , Eritropoyetina/genética , Femenino , Humanos , Estimación de Kaplan-Meier , Células MCF-7 , Ratones Endogámicos C57BL , Ratones Desnudos , Persona de Mediana Edad , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Unión Proteica/efectos de los fármacos , Receptor EphB4/metabolismo , Receptores de Eritropoyetina/genética , Receptores de Eritropoyetina/metabolismo , Proteínas Recombinantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Adulto Joven
13.
J Cereb Blood Flow Metab ; 24(2): 224-36, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14747749

RESUMEN

Studies of gene expression changes after cerebral ischemia can provide novel insight into ischemic pathophysiology. Here we describe application of restriction-mediated differential display to screening for differentially expressed genes after focal cerebral ischemia. This method combines the nonredundant generation of biotin-labeled fragment sets with the excellent resolution of direct blotting electrophoresis, reliable fragment recovery, and a novel clone selection strategy. Using the filament model in mouse with 90 minutes MCA occlusion followed by 2, 6, and 20 hours reperfusion, we have compared gene expression in sham-operated animals to both the ipsi- and contralateral forebrain hemisphere of ischemic mice. Our screening method has resulted in the identification of 70 genes differentially regulated after transient middle cerebral artery occlusion (MCAO), several of which represent unknown clones. We have identified many of the previously published regulated genes, lending high credibility to our method. Surprisingly, we detected a high degree of correspondent regulation of genes in the nonischemic hemisphere. A high percentage of genes coding for proteins in the respiratory chain was found to be up-regulated after ischemia, potentially representing a new mechanism involved in counteracting energy failure or radical generation in cerebral ischemia. One particularly interesting gene, whose upregulation by ischemia has not been described before, is pip92; this gene shows a rapid and long-lasting induction after cerebral ischemia. Here we demonstrate that pip92 induces cell death in primary neurons and displays several hallmarks of pro-apoptotic activity upon overexpression, supporting the notion that we have identified a novel pathophysiological player in cerebral ischemia. In summary, restriction-mediated differential display has proven its suitability for screening complex samples such as brain to reliably identify regulated genes, which can uncover novel pathophysiological mechanisms.


Asunto(s)
Apoptosis , Isquemia Encefálica/fisiopatología , Perfilación de la Expresión Génica/métodos , Proteínas/metabolismo , Animales , Células COS , Fragmentación del ADN , Regulación de la Expresión Génica , Proteínas Inmediatas-Precoces , Infarto de la Arteria Cerebral Media , Ratones , Neuronas/citología , Neuronas/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas/genética , Factores de Tiempo
14.
Brain Res Mol Brain Res ; 124(1): 20-8, 2004 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-15093682

RESUMEN

Cerebral ischemia induces transcriptional changes in a number of pathophysiologically important genes. Here we have systematically studied gene expression changes in the cortex after 150 min of focal cortical ischemia and 2 and 6 h reperfusion in the mouse by a fragment display technique (restriction-mediated differential display, RMDD). We identified 57 transcriptionally altered genes, of which 46 were known genes, and 11 unknown sequences. Of note, 14% of the regulated genes detected at 2 h reperfusion time were co-regulated in the contralateral cortex. Four genes were verified to be upregulated by quantitative PCR. These were Metallothionein-II (mt2), Receptor (calcitonin)-activity modifying protein 2 (ramp2), Mitochondrial phosphoprotein 65 (MIPP65), and the transcription elongation factor B2/elongin B (tceb). We could identify several genes that are known to be induced by cerebral ischemia, such as the metallothioneins and c-fos. Many of the genes identified provide hints to potential new mechanisms in ischemic pathophysiology. We discuss the identity of the regulated genes in view of their possible usefulness for pharmacological intervention in cerebral ischemia.


Asunto(s)
Regulación de la Expresión Génica , Ataque Isquémico Transitorio/genética , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Animales , Northern Blotting/métodos , Enfermedades de las Arterias Carótidas/complicaciones , Enfermedades de las Arterias Carótidas/genética , Clonación Molecular , Biología Computacional/métodos , Perfilación de la Expresión Génica , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/genética , Ataque Isquémico Transitorio/etiología , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa/métodos , ARN/biosíntesis , Factores de Tiempo
15.
Exp Transl Stroke Med ; 6(1): 3, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24528872

RESUMEN

BACKGROUND AND PURPOSE: Both the immobilization of the unaffected arm combined with physical therapy (forced arm use, FAU) and voluntary exercise (VE) as model for enriched environment are promising approaches to enhance recovery after stroke. The genomic mechanisms involved in long-term plasticity changes after different means of rehabilitative training post-stroke are largely unexplored. The present investigation explored the effects of these physical therapies on behavioral recovery and molecular markers of regeneration after experimental ischemia. METHODS: 42 Wistar rats were randomly treated with either forced arm use (FAU, 1-sleeve plaster cast onto unaffected limb at 8/10 days), voluntary exercise (VE, connection of a freely accessible running wheel to cage), or controls with no access to a running wheel for 10 days starting at 48 hours after photothrombotic stroke of the sensorimotor cortex. Functional outcome was measured using sensorimotor test before ischemia, after ischemia, after the training period of 10 days, at 3 and 4 weeks after ischemia. Global gene expression changes were assessed from the ipsi- and contralateral cortex and the hippocampus. RESULTS: FAU-treated animals demonstrated significantly improved functional recovery compared to the VE-treated group. Both were superior to cage control. A large number of genes are altered by both training paradigms in the ipsi- and contralateral cortex and the hippocampus. Overall, the extent of changes observed correlated well with the functional recovery obtained. One category of genes overrepresented in the gene set is linked to neuronal plasticity processes, containing marker genes such as the NMDA 2a receptor, PKC ζ, NTRK2, or MAP 1b. CONCLUSIONS: We show that physical training after photothrombotic stroke significantly and permanently improves functional recovery after stroke, and that forced arm training is clearly superior to voluntary running training. The behavioral outcomes seen correlate with patterns and extent of gene expression changes in all brain areas examined. We propose that physical training induces a fundamental change in plasticity-relevant gene expression in several brain regions that enables recovery processes. These results contribute to the debate on optimal rehabilitation strategies, and provide a valuable source of molecular entry points for future pharmacological enhancement of recovery.

16.
Front Cell Neurosci ; 8: 464, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25653590

RESUMEN

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is an incurable fatal motoneuron disease with a lifetime risk of approximately 1:400. It is characterized by progressive weakness, muscle wasting, and death ensuing 3-5 years after diagnosis. Granulocyte-colony stimulating factor (G-CSF) is a drug candidate for ALS, with evidence for efficacy from animal studies and interesting data from pilot clinical trials. To gain insight into the disease mechanisms and mode of action of G-CSF, we performed gene expression profiling on isolated lumbar motoneurons from SOD1(G93A) mice, the most frequently studied animal model for ALS, with and without G-CSF treatment. RESULTS: Motoneurons from SOD1(G93A) mice present a distinct gene expression profile in comparison to controls already at an early disease stage (11 weeks of age), when treatment was initiated. The degree of deregulation increases at a time where motor symptoms are obvious (15 weeks of age). Upon G-CSF treatment, transcriptomic deregulations of SOD1(G93A) motoneurons were notably restored. Discriminant analysis revealed that SOD1 mice treated with G-CSF has a transcriptom close to presymptomatic SOD1 mice or wild type mice. Some interesting genes modulated by G-CSF treatment relate to neuromuscular function such as CCR4-NOT or Prss12. CONCLUSIONS: Our data suggest that G-CSF is able to re-adjust gene expression in symptomatic SOD1(G93A) motoneurons. This provides further arguments for G-CSF as a promising drug candidate for ALS.

17.
PLoS One ; 5(5): e10737, 2010 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-20505770

RESUMEN

BACKGROUND: We have previously identified Semaphorin 6a (Sema6A) as an upregulated gene product in a gene expression screen in cortical ischemia [1]. Semaphorin 6a was regulated during the recovery phase following ischemia in the cortex. Semaphorin 6a is a member of the superfamily of semaphorins involved in axon guidance and other functions. We hypothesized that the upregulation indicates a crucial role of this molecule in post-stroke rewiring of the brain. Here we have tested this hypothesis by overexpressing semaphorin 6a in the cortex by microinjection of a modified AAV2-virus. A circumscribed cortical infarct was induced, and the recovery of rats monitored for up to 4 weeks using a well-established test battery (accelerated rotarod training paradigm, cylinder test, adhesive tape removal). We observed a significant improvement in post-ischemic recovery of animals injected with the semaphorin 6a virus versus animals treated with a control virus. We conclude that semaphorin 6a overexpressed in the cortex enhances recovery after cerebral ischemia. Semaphorin 6a may represent a novel therapeutic candidate for the treatment of chronic stroke.


Asunto(s)
Isquemia Encefálica/fisiopatología , Actividad Motora/fisiología , Condicionamiento Físico Animal , Recuperación de la Función/fisiología , Semaforinas/metabolismo , Animales , Isquemia Encefálica/metabolismo , Línea Celular , Dependovirus/genética , Humanos , Desempeño Psicomotor , Ratas , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/fisiopatología
18.
J Cereb Blood Flow Metab ; 29(3): 585-95, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19116637

RESUMEN

Hemoglobin is the major protein in red blood cells and transports oxygen from the lungs to oxygen-demanding tissues, like the brain. Mechanisms that facilitate the uptake of oxygen in the vertebrate brain are unknown. In invertebrates, neuronal hemoglobin serves as intracellular storage molecule for oxygen. Here, we show by immunohistochemistry that hemoglobin is specifically expressed in neurons of the cortex, hippocampus, and cerebellum of the rodent brain, but not in astrocytes and oligodendrocytes. The neuronal hemoglobin distribution is distinct from the neuroglobin expression pattern on both cellular and subcellular levels. Probing for low oxygen levels in the tissue, we provide evidence that hemoglobin alpha-positive cells in direct neighborhood with hemoglobin alpha-negative cells display a better oxygenation than their neighbors and can be sharply distinguished from those. Neuronal hemoglobin expression is upregulated by injection or transgenic overexpression of erythropoietin and is accompanied by enhanced brain oxygenation under physiologic and hypoxic conditions. Thus we provide a novel mechanism for the neuroprotective actions of erythropoietin under ischemic-hypoxic conditions. We propose that neuronal hemoglobin expression is connected to facilitated oxygen uptake in neurons, and hemoglobin might serve as oxygen capacitator molecule.


Asunto(s)
Cerebelo/metabolismo , Corteza Cerebral/metabolismo , Hemoglobinas/biosíntesis , Hipocampo/metabolismo , Neuronas/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Hipoxia de la Célula , Células Cultivadas , Cerebelo/citología , Cerebelo/efectos de los fármacos , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Electroforesis en Gel Bidimensional , Eritropoyetina/genética , Eritropoyetina/farmacología , Femenino , Hipocampo/citología , Hipocampo/efectos de los fármacos , Espectrometría de Masas , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/efectos de los fármacos , Nitroimidazoles/farmacología , Oxígeno/metabolismo , Ratas , Ratas Wistar , Proteínas Recombinantes , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Especificidad de la Especie
19.
J Proteome Res ; 6(3): 1198-208, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17330951

RESUMEN

On the basis of its inhibition by SB216763, we identified the multifunctional enzyme Glycogen Synthase Kinase 3beta (GSK3beta) as a central regulator for differentiation and cell survival of adult neural stem cells. Detected by proteomic approaches, members of the Wnt/beta-catenin signaling pathway appear to participate in enhanced neuronal differentiation and activated transcription of beta-catenin target genes during GSK3beta inhibition, associated with decreased apoptosis.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Ventrículos Cerebrales/citología , Glucógeno Sintasa Quinasa 3/fisiología , Neuronas/citología , Células Madre/citología , Animales , Apoptosis , Electroforesis en Gel Bidimensional , Glucógeno Sintasa Quinasa 3 beta , Neuronas/enzimología , Proteómica/métodos , Ratas , Células Madre/enzimología , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
20.
Exp Neurol ; 200(1): 135-52, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16530183

RESUMEN

Cerebral ischemia evokes changes in gene expression time-dependently after the ischemic event. Most studies on transcriptional changes following ischemia have centered on relatively early postischemic time points, and detected multiple genes relevant to neuronal cell death. However, functional outcome after ischemia depends critically on adaptations of the postischemic brain. Plasticity may derive from network-inherent changes, or from the formation of new nerve cells in the CNS. We have screened for gene expression changes up to 3 weeks following a limited photothrombotic cortical insult in the rat sensorimotor cortex by using the sensitive restriction-mediated differential display (RMDD) technique. A high number of genes were detected as induced at early or intermediate time points in the ipsi- and contralateral cortex (6 and 48 h). Unexpectedly, at the late time point examined (3 weeks), we still detected 40 genes that were changed in their expression. We further characterized the expression of two genes linked to neurogenesis (nestin and stathmin), and two genes likely involved in reconfiguring neuronal networks (semaphorin VIa and synaptotagmin IV). Conclusively, our data highlight the degree of long-term transcriptional changes in the cortex after ischemia, and provide insight into functional pathways of relevance for compensatory recovery mechanisms in neural networks.


Asunto(s)
Química Encefálica/genética , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Corteza Cerebral/metabolismo , Perfilación de la Expresión Génica , Activación Transcripcional/fisiología , Animales , Química Encefálica/fisiología , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/fisiología , Ratas , Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA