Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Clin Sci (Lond) ; 131(10): 917-934, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28473472

RESUMEN

Nuclear receptors (NRs) are cellular proteins, which upon ligand activation, act to exert regulatory control over transcription and subsequent expression. Organized via systemic classification into seven subfamilies, NRs partake in modulating a vast expanse of physiological functions essential for maintenance of life. NRs display particular characteristics towards ubiquitination, the process of addition of specific ubiquitin tags at appropriate locations. Orchestrated through groups of enzymes harboring a diverse array of specialized structural components, the ubiquitination process emphatically alters the fate or downstream effects of NRs. Such influence is especially prominent in transcriptional processes such as promoter clearing for optimization and degradation pathways eliminating or recycling targeted proteins. Ultimately, the ubiquitination of NRs carries significant implications in terms of generating pathological clinical manifestations. Increasing evidence from studies involving patients and disease models suggests a role for ubiquitinated NRs in virtually every organ system. This supports the broad repertoire of roles that NRs play in the body, including modulatory conductors, facilitators, responders to external agents, and critical constituents for pharmacological or biological interventions. This review aims to cover relevant background and mechanisms of NRs and ubiquitination, with a focus towards elucidating subsequent pathophysiology and therapeutics in clinical disorders encompassing such ubiquitinated NRs.


Asunto(s)
Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Regulación de la Expresión Génica , Humanos , Receptores Citoplasmáticos y Nucleares/genética , Transducción de Señal , Ubiquitina/metabolismo , Ubiquitinación
2.
IUBMB Life ; 68(7): 504-15, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27240871

RESUMEN

Coregulators span a broad and extensive domain in modulating cellular transcriptional activity. Studies have established a dynamic role for such coregulators in various endocrine cancers. Steroid hormone receptors (SHRs) play a pivotal role in such endocrine cancers, and interact abundantly with transcriptional coregulators in altering gene expression. Several families of coregulators have implications in propagating the development, progression and invasion of breast, prostate, and other hormone-responsive cancers. This mini-review aims to discuss different classes of coregulators involved in endocrine cancers and highlight unique information regarding each family with relevance to mechanism, intervention, and novel directions being investigated. © 2016 IUBMB Life, 68(7):504-515, 2016.


Asunto(s)
Neoplasias de las Glándulas Endocrinas/genética , Regulación Neoplásica de la Expresión Génica/genética , ARN Mensajero/genética , Receptores de Esteroides/genética , Proteínas Portadoras/genética , Neoplasias de las Glándulas Endocrinas/patología , Humanos , Coactivador 1 de Receptor Nuclear/genética , Proteínas Inhibidoras de STAT Activados/genética , ARN Mensajero/biosíntesis , Ubiquitina-Proteína Ligasas/genética
3.
J Mol Recognit ; 26(2): 92-103, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23334917

RESUMEN

Allostery plays a key role in dictating the stoichiometry and thermodynamics of multi-protein complexes driving a plethora of cellular processes central to health and disease. Herein, using various biophysical tools, we demonstrate that although Sos1 nucleotide exchange factor and Gab1 docking protein recognize two non-overlapping sites within the Grb2 adaptor, allostery promotes the formation of two distinct pools of Grb2-Sos1 and Grb2-Gab1 binary signaling complexes in concert in lieu of a composite Sos1-Grb2-Gab1 ternary complex. Of particular interest is the observation that the binding of Sos1 to the nSH3 domain within Grb2 sterically blocks the binding of Gab1 to the cSH3 domain and vice versa in a mutually exclusive manner. Importantly, the formation of both the Grb2-Sos1 and Grb2-Gab1 binary complexes is governed by a stoichiometry of 2:1, whereby the respective SH3 domains within Grb2 homodimer bind to Sos1 and Gab1 via multivalent interactions. Collectively, our study sheds new light on the role of allostery in mediating cellular signaling machinery.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Proteína Adaptadora GRB2/química , Proteína SOS1/química , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales/genética , Regulación Alostérica , Secuencias de Aminoácidos , Sitios de Unión , Escherichia coli/genética , Proteína Adaptadora GRB2/genética , Humanos , Cinética , Ligandos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Multimerización de Proteína , Estructura Secundaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteína SOS1/genética , Termodinámica
4.
J Proteome Res ; 11(6): 3053-67, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22559202

RESUMEN

Quantitative proteomics analysis of cortical samples of 10 Alzheimer's disease (AD) brains versus 10 normally aged brains was performed by following the accurate mass and time tag (AMT) approach with the high resolution LTQ Orbitrap mass spectrometer. More than 1400 proteins were identified and quantitated. A conservative approach of selecting only the consensus results of four normalization methods was suggested and used. A total of 197 proteins were shown to be significantly differentially abundant (p-values <0.05, corrected for multiplicity of testing) in AD versus control brain samples. Thirty-seven of these proteins were reported as differentially abundant or modified in AD in previous proteomics and transcriptomics publications. The rest to the best of our knowledge are new. Mapping of the discovered proteins with bioinformatic tools revealed significant enrichment with differentially abundant proteins of pathways and processes known to be important in AD, including signal transduction, regulation of protein phosphorylation, immune response, cytoskeleton organization, lipid metabolism, energy production, and cell death.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Proteoma/metabolismo , Factores de Transcripción/metabolismo , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Cromatografía Liquida , Redes Reguladoras de Genes , Humanos , Masculino , Proteoma/genética , Proteoma/aislamiento & purificación , Proteómica , Reproducibilidad de los Resultados , Espectrometría de Masas en Tándem , Factores de Transcripción/genética , Factores de Transcripción/aislamiento & purificación
5.
Methods ; 48(1): 14-8, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19250969

RESUMEN

Mismatch repair corrects biosynthetic errors generated during DNA replication, whose deficiency causes a mutator phenotype and directly underlies hereditary non-polyposis colorectal cancer and sporadic cancers. Because of remarkably high conservation of the mismatch repair machinery between the budding yeast (Saccharomyces cerevisiae) and humans, the study of mismatch repair in yeast has provided tremendous insights into the mechanisms of this repair pathway in humans. In addition, yeast cells possess an unbeatable advantage over human cells in terms of the easy genetic manipulation, the availability of whole genome deletion strains, and the relatively low cost for setting up the system. Although many components of eukaryotic mismatch repair have been identified, it remains unclear if additional factors, such as DNA helicase(s) and redundant nuclease(s) besides EXO1, participate in eukaryotic mismatch repair. To facilitate the discovery of novel mismatch repair factors, we developed a straightforward in vitro cell-free repair system. Here, we describe the practical protocols for preparation of yeast cell-free nuclear extracts and DNA mismatch substrates, and the in vitro mismatch repair assay. The validity of the cell-free system was confirmed by the mismatch repair deficient yeast strain (Deltamsh2) and the complementation assay with purified yeast MSH2-MSH6.


Asunto(s)
Reparación de la Incompatibilidad de ADN/genética , Reparación del ADN , ADN de Hongos/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Núcleo Celular/metabolismo , Sistema Libre de Células/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/metabolismo , Regulación Fúngica de la Expresión Génica , Prueba de Complementación Genética , Proteína 2 Homóloga a MutS/metabolismo , Reproducibilidad de los Resultados , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/análisis , Proteínas de Saccharomyces cerevisiae/aislamiento & purificación , Especificidad por Sustrato
6.
Epilepsy Res ; 81(2-3): 161-6, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18603412

RESUMEN

Status epilepticus (SE) induces a number of events leading to programmed cell death (PCD). The aim of our work is to study the time sequence of activation of different factors in experimental SE (intraperitoneal kainic acid (KA) model). We studied ceramide, a known mediator of apoptosis in multiple models, sphingomyelinases (SMases), enzymes that break down sphingomyelin and increase ceramide thus leading to apoptosis in many models, Bcl(2), Bax, and caspase-3. SE induced a sustained ceramide increase starting 2h after kainic acid injection followed by an increase in Bax protein at 6 and 12h, and the appearance of caspase-3-activated fragment (caspase-3a) immunostaining and TUNEL positivity at 12h. Status epilepticus also induced an increase in acidic and neutral sphingomyelinases that preceded (acidic sphingomyelinase) and parallelled (acidic and neutral sphingomyelinase) the increases in ceramide. These data suggest that, in this model, Bax is activated early in the process and that its increase is sustained till 12h after kainic acid injection which is the time of first appearance of caspase-3 activation and TUNEL positivity, and that SMases contribute to increases in ceramide levels during and after status epilepticus.


Asunto(s)
Caspasa 3/metabolismo , Ceramidas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Estado Epiléptico/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Animales , Anticonvulsivantes/uso terapéutico , Muerte Celular/efectos de los fármacos , Diazepam/uso terapéutico , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Etiquetado Corte-Fin in Situ/métodos , Ácido Kaínico , Ratas , Ratas Sprague-Dawley , Estado Epiléptico/inducido químicamente , Estado Epiléptico/tratamiento farmacológico , Estado Epiléptico/fisiopatología , Factores de Tiempo
7.
Brain Dev ; 30(8): 513-9, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18295995

RESUMEN

Ceramide is known to induce programmed cell death (PCD) in neural and non-neural tissues and to increase after kainic acid (KA) status epilepticus (SE). Ceramide increases have been shown to depend on NMDA receptor activation in the KA model, but these changes have not been studied in the lithium pilocarpine (LiPC) model. Thus, the purpose of this study was to determine if hippocampal ceramide levels increase after LiPC induced SE and if NMDA receptor blockade prevents PCD and any such ceramide increases. We found that LiPC induced SE resulted in ceramide increases and DNA fragmentation in the hippocampus of adult, P21, and P7 rats. The administration of MK-801, the NMDA receptor antagonist, in adults, 15min prior to pilocarpine, prevented ceramide increases, and DNA fragmentation.


Asunto(s)
Muerte Celular/fisiología , Ceramidas/metabolismo , Litio/farmacología , Pilocarpina/farmacología , Receptores de N-Metil-D-Aspartato/metabolismo , Convulsiones , Animales , Modelos Animales de Enfermedad , Maleato de Dizocilpina/metabolismo , Antagonistas de Aminoácidos Excitadores/metabolismo , Femenino , Humanos , Etiquetado Corte-Fin in Situ , Ácido Kaínico/farmacología , Masculino , Ratas , Ratas Sprague-Dawley , Convulsiones/inducido químicamente , Convulsiones/metabolismo
8.
Mol Neurobiol ; 55(9): 7187-7200, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29388081

RESUMEN

Angelman syndrome (AS) is a complex genetic disorder that affects the nervous system. AS affects an estimated 1 in 12,000 to 20,000 individuals. Characteristic features of AS includes developmental delay or intellectual disability, severe speech impairment, seizures, small head size (microcephaly), and problems with movement and balance (ataxia). AS individuals usually have microdeletion of the maternal copy of 15q11.2-15q13 region of chromosome 15. The E6-associated protein (E6AP, an E3 ubiquitin protein ligase enzyme) is encoded by the gene UBE3A, which is located in this region, and it has been shown that deregulation of E6AP gives rise to AS and neuropathology of autism spectrum disorders (ASDs) (e.g., autism and Rett syndromes). We have shown that E6AP also acts as a coactivator of the estrogen receptor (ER). ER is a ligand-induced transcription factor that exerts potent and wide-ranging effects on the developing brain. Furthermore, the expression pattern of ER in the brain overlaps with that of E6AP. Up till now, all the published studies have examined the role of the ubiquitin-protein ligase activity of E6AP in the development of AS, and it is not known what role the newly discovered coactivation functions of E6AP and ER plays in the pathology of AS. Here, we demonstrate that E6AP and ER co-immunoprecipitate and are in the same protein complex in neuronal cells (Neuro2a). In addition, both colocalize in nuclear and cytoplasmic compartments of the mouse hippocampal neurons and Neuro2a cells. Moreover, we identified a novel E6AP and ER direct transcriptional regulation of a gene Cyp26b1 known to be involved in learning and memory processes. This transcriptional regulation involves recruitment of E6AP and ER to a newly discovered functional estrogen response element (ERE) located at the Cyp26b1 gene promoter and is associated with transcription permissive epigenetic events leading to increase of active transcription of the gene in neurons upon estrogen treatment. This novel transcriptional regulation was also validated in the AS mouse model where E6AP expression is abrogated in the mouse brain. In fact, Cyp26b1 expression is decreased by 31% in AS mice versus age-matched control (Ctrl) mice hippocampi. Also, retinoic acid transcriptional signaling was shown to be amplified as evidenced by specific increased Rarß and decreased Erbb4 mRNA expression in AS mice versus Ctrl mice hippocampi. These transcript level changes were also supported by the same trend of changes at the protein level. Collectively, our data present a proof of principle that the transcriptional coactivation function of E6AP may have a crucial role in the pathobiology of AS. This function, yet to be thoroughly investigated, reveals the possibility of harnessing the antagonistic estrogen-retinoic acid transcriptional signaling crosstalk and potentially other unknown effectors for the investigation of important possible targets as putative novel treatment modalities and venues for reversing neurological manifestations in AS and related syndromes like ASDs.


Asunto(s)
Síndrome de Angelman/genética , Estrógenos/metabolismo , Neuronas/metabolismo , Transcripción Genética , Tretinoina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Síndrome de Angelman/patología , Animales , Secuencia de Bases , Núcleo Celular/metabolismo , Epigénesis Genética/efectos de los fármacos , Células HeLa , Humanos , Ratones Endogámicos C57BL , Modelos Biológicos , Neuronas/patología , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Estrógenos/metabolismo , Elementos de Respuesta/genética , Ácido Retinoico 4-Hidroxilasa/genética , Ácido Retinoico 4-Hidroxilasa/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
9.
Mol Neurobiol ; 55(2): 1714-1724, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28214999

RESUMEN

E6ap is a known transcriptional coregulator for estrogen receptor alpha (Er, Erα) in the presence of estrogen. Protein kinase A (PKA) contains two regulatory subunits derived from four genes. Recent evidence demonstrates that PKA regulates E6ap activity. Data generated in our lab indicated estrogen dependent regulation of Pkar2a levels. Our project sets to investigate a possible feedback mechanism constituting of Erα and E6ap transcriptional regulation of Pkar2a expression. Western blot evaluated protein regulation correlations with E2 in mouse neuroblastoma lines. Bioinformatics detected estrogen response element (ERE) sequences. quantitative polymerase chain reaction (qPCR) validated the western blot results. ERE oligonucleotides were synthesized. Reporter gene transcriptional activity was evaluated via Luciferase assay output. Electromobility shift assay (EMSA) assessed direct binding between Erα relevant sequences. Chromatin immunoprecipitation (ChIP) and Re-ChIP were conducted in quantifying protein complex recruitment levels. Pkar2a protein expression directly correlated with E2, and four putative ERE sequences were identified. Pkar2a mRNA expression reverted to baseline with either E2 or E6ap absent. In the presence of E2, ERE-1 and ERE-4 possessed Luciferase reporter gene transcriptional capabilities. ERE-1 portrayed band shifts, representing direct binding to Erα with E2 supplementation. With E2, ERE-1 significantly enhanced Erα and E6ap recruitment levels to the Pkar2a promoter. Pkar2a is directly regulated by Erα and E6ap in the presence of estrogen stimulus. This work indicates a feedback mechanism in the interplay between PKA and E6ap, which may prove crucial for the role of both proteins in cancers and neurogenetic diseases like Angelman syndrome.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Neuroblastoma/metabolismo , Neuronas/metabolismo , Receptores de Estrógenos/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular Tumoral , Estradiol/farmacología , Ratones , Neuronas/efectos de los fármacos , Regiones Promotoras Genéticas , Elementos de Respuesta/fisiología
10.
Sci Rep ; 8(1): 8423, 2018 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-29849033

RESUMEN

Potentially pathogenic alterations have been identified in individuals with autism spectrum disorders (ASDs) within a variety of key neurodevelopment genes. While this hints at a common ASD molecular etiology, gaps persist in our understanding of the neurodevelopmental mechanisms impacted by genetic variants enriched in ASD patients. Induced pluripotent stem cells (iPSCs) can model neurodevelopment in vitro, permitting the characterization of pathogenic mechanisms that manifest during corticogenesis. Taking this approach, we examined the transcriptional differences between iPSC-derived cortical neurons from patients with idiopathic ASD and unaffected controls over a 135-day course of neuronal differentiation. Our data show ASD-specific misregulation of genes involved in neuronal differentiation, axon guidance, cell migration, DNA and RNA metabolism, and neural region patterning. Furthermore, functional analysis revealed defects in neuronal migration and electrophysiological activity, providing compelling support for the transcriptome analysis data. This study reveals important and functionally validated insights into common processes altered in early neuronal development and corticogenesis and may contribute to ASD pathogenesis.


Asunto(s)
Trastorno Autístico/genética , Trastorno Autístico/patología , Perfilación de la Expresión Génica , Neuronas/metabolismo , Adolescente , Señalización del Calcio , Diferenciación Celular , Movimiento Celular , Niño , Preescolar , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Masculino , Neuronas/patología , Sinapsis/patología , Adulto Joven
11.
Artículo en Inglés | MEDLINE | ID: mdl-17901003

RESUMEN

A rapid, reproducible and accurate high-performance liquid chromatographic (HPLC) method for the quantitative determination of sphingomyelin in rat brain was developed and validated using normal-phase silica gel column, acetonitrile-methanol-water (65:18:17 (v/v)) at a flow rate of 1 ml/min, isocratic elution, UV detection at 207 nm and 1,2-dimyristoyl-sn-glycero-3-phosphocholine as an internal standard. Total run time was 10.0 min. The calibration curve was linear over the range of 0.025-0.4 mg/ml sphingomyelin (R2>0.99). The intra-day coefficient of variation ranged from 1.4% to 2.2%. The average inter-day coefficient of variation over a period of 4 days was 3.1%. The practical limit of detection was 0.005 mg/ml with a quantification limit of 0.01 mg/ml.


Asunto(s)
Química Encefálica , Cromatografía Líquida de Alta Presión/métodos , Esfingomielinas/análisis , Animales , Dimiristoilfosfatidilcolina/normas , Ratas , Estándares de Referencia , Sensibilidad y Especificidad
12.
Mol Neurobiol ; 54(2): 1301-1313, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-26843107

RESUMEN

The tumor necrosis factor receptor-associated factors (TRAFs) have been classically described as adaptor proteins that function as solely cytosolic signaling intermediates for the TNF receptor superfamily, Toll-like receptors (TLRs), NOD, like receptors (NLRs), cytokine receptors, and others. In this study, we show for the first time that TRAFs are present within the cytoplasm and nucleus of Neuro2a cells and primary cortical neurons, and that TRAF2 and TRAF3 translocate into the nucleus within minutes of CD40L stimulation. Analysis of the transcriptional regulatory potential of TRAFs by luciferase assay revealed that each of the TRAFs differentially functions as a transcriptional activator or repressor in a cell-specific manner. Interestingly, ChIP-qPCR data demonstrate that TRAFs 2/3, p65, and pRNAPol II form part of a transcriptional complex on the Icam-1 gene promoter upon CD40L stimulation. We further determined that TRAF2 recruitment to the nucleus is critical for the ubiquitination of H2b, a transcription permissive epigenetic modification. Our findings demonstrate for the first time that TRAFs 2/3 participate in the formation of a CD40L-induced transcriptional complex in neuronal cells.


Asunto(s)
Antígenos CD40/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas de Transporte Nucleocitoplasmático/metabolismo , ARN Polimerasa II/metabolismo , Factor 2 Asociado a Receptor de TNF/metabolismo , Factor 3 Asociado a Receptor de TNF/metabolismo , Animales , Antígenos CD40/análisis , Línea Celular Tumoral , Núcleo Celular/química , Núcleo Celular/metabolismo , Citoplasma/química , Citoplasma/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas de Neoplasias/análisis , Proteínas de Transporte Nucleocitoplasmático/análisis , ARN Polimerasa II/análisis , Factor 2 Asociado a Receptor de TNF/análisis , Factor 3 Asociado a Receptor de TNF/análisis , Activación Transcripcional/fisiología
13.
J Med Liban ; 54(4): 200-4, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17330372

RESUMEN

Because magnesium has antiseizure effects in some animal models of epilepsy, and possible neuroprotective effects in some models of neuronal injury, we aimed to investigate its effects in the kainic acid (KA) model of status epilepticus (SE) in prepubescent rats. This age was chosen because it is a common age for onset of epilepsy and of SE in humans. Three groups of P35 rats were studied: Group I (MgKA) received magnesium sulfate MgSO4 (270 mg/kg then 27 mg/kg every 20 minutes for 5 hours) and 10 mg/kg KA. Group II (KA) received saline instead of MgSO4 and 10 mg/kg KA. Group III (control) received saline injections only. The dose we used has been shown previously to have anticonvulsant activity in another seizure model. Rats were recorded for their acute behavioral seizures directly after KA, and underwent the handling and Morris Water Maze (MWM) tests on P96-97 and P102-106 respectively. The MgKA and the KA groups did not differ in their acute seizures and both showed similar histologic lesions in CA3/CA4 and CA1 hippocampal subfields, and were more aggressive on the handling test than control rats. The MgKA group took more time to reach the platform in MWM than controls, while the KA group scores were intermediate between the two groups. Using the dose of 540 mg/kg MgSO4 and 54 mg/kg every 20 min showed the similar result of lack of protection against impairment in long-term memory. We conclude that (1) Magnesium did not manifest acute behavioral antiseizure effects in the KA P35 model of SE. (2) Magnesium did not prevent the tested long-term behavioral and histological consequences of SE in this model.


Asunto(s)
Anticonvulsivantes/farmacología , Sulfato de Magnesio/farmacología , Estado Epiléptico/tratamiento farmacológico , Animales , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad , Esquema de Medicación , Agonistas de Aminoácidos Excitadores/farmacología , Hipocampo/patología , Ácido Kaínico/farmacología , Ratas , Ratas Sprague-Dawley , Estado Epiléptico/inducido químicamente
14.
Artículo en Inglés | MEDLINE | ID: mdl-27882265

RESUMEN

Since the creation of induced Pluripotent Stem Cells (iPSCs) ten years ago, hundreds of publications have demonstrated their considerable impact on disease modeling and therapy. In this commentary, we will summarize key milestones, benefits and challenges in the iPSC field. Furthermore, we will highlight blood as an effective and easily accessible source for patient-specific iPSCs derivation in the context of work done in our laboratory and others.

15.
Stem Cells Dev ; 25(11): 848-60, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27059084

RESUMEN

Human bone marrow multipotent mesenchymal stromal cell (hMSC) number decreases with aging. Subpopulations of hMSCs can differentiate into cells found in bone, vasculature, cartilage, gut, and other tissues and participate in their repair. Maintaining throughout adult life such cell subpopulations should help prevent or delay the onset of age-related degenerative conditions. Low oxygen tension, the physiological environment in progenitor cell-rich regions of the bone marrow microarchitecture, stimulates the self-renewal of marrow-isolated adult multilineage inducible (MIAMI) cells and expression of Sox2, Nanog, Oct4a nuclear accumulation, Notch intracellular domain, notch target genes, neuronal transcriptional repressor element 1 (RE1)-silencing transcription factor (REST), and hypoxia-inducible factor-1 alpha (HIF-1α), and additionally, by decreasing the expression of (i) the proapoptotic proteins, apoptosis-inducing factor (AIF) and Bak, and (ii) senescence-associated p53 expression and ß-galactosidase activity. Furthermore, low oxygen increases canonical Wnt pathway signaling coreceptor Lrp5 expression, and PI3K/Akt pathway activation. Lrp5 inhibition decreases self-renewal marker Sox2 mRNA, Oct4a nuclear accumulation, and cell numbers. Wortmannin-mediated PI3K/Akt pathway inhibition leads to increased osteoblastic differentiation at both low and high oxygen tension. We demonstrate that low oxygen stimulates a complex signaling network involving PI3K/Akt, Notch, and canonical Wnt pathways, which mediate the observed increase in nuclear Oct4a and REST, with simultaneous decrease in p53, AIF, and Bak. Collectively, these pathway activations contribute to increased self-renewal with concomitant decreased differentiation, cell cycle arrest, apoptosis, and/or senescence in MIAMI cells. Importantly, the PI3K/Akt pathway plays a central mechanistic role in the oxygen tension-regulated self-renewal versus osteoblastic differentiation of progenitor cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Células de la Médula Ósea/citología , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Autorrenovación de las Células/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Oxígeno/farmacología , Transducción de Señal/efectos de los fármacos , Adulto , Apoptosis/genética , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Diferenciación Celular/genética , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Autorrenovación de las Células/genética , Separación Celular , Senescencia Celular/genética , Niño , Preescolar , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Masculino , Modelos Biológicos , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Receptores Notch/metabolismo , Transducción de Señal/genética , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo
16.
Brain Res Dev Brain Res ; 157(1): 98-102, 2005 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-15939090

RESUMEN

Ten-day-old rat pups (P10) subjected to acute hypoxia (down to 4% O2) had as adults increased aggression (handling test), memory impairment (water maze test), and decreased CA1 cell counts. Pups subjected to chronic hypoxia (10% O2 from P0 to P21) had increased aggression, hyperactivity (open-field test), and decreased CA1 cell counts. Chronic hypoxia with superimposed acute hypoxia resulted in consequences that were not different from those of chronic hypoxia.


Asunto(s)
Conducta Animal/fisiología , Hipocampo , Hipoxia/patología , Hipoxia/fisiopatología , Tiempo , Análisis de Varianza , Animales , Animales Recién Nacidos , Recuento de Células/métodos , Muerte Celular/fisiología , Enfermedad Crónica , Ensayo de Inmunoadsorción Enzimática , Eritropoyetina/sangre , Conducta Exploratoria/fisiología , Hipocampo/crecimiento & desarrollo , Hipocampo/patología , Hipocampo/fisiopatología , Hipoxia/sangre , Etiquetado Corte-Fin in Situ/métodos , Aprendizaje por Laberinto/fisiología , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
17.
Mol Neurobiol ; 49(2): 827-39, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24091829

RESUMEN

E6-Associated Protein (E6AP), the founding member of the HECT (Homologus to E6AP C terminus) family of ubiquitin ligases, has been gaining increased attention from the scientific community. In addition to its ubiquitin ligase function, our laboratory has also identified steroid hormone receptor transcriptional coactivation as yet another essential function of this protein. Furthermore, it has been established that E6AP has a role in numerous diseases including cancers and neurological syndromes. In this review, we delineate genetic and biochemical knowledge of E6AP and we focus on its role in the pathobiology of neuro-developmental and neuro-aging diseases; bringing to light important gaps of knowledge related to the involvement of its well-studied ligase function versus the much less studied nuclear receptor transcriptional coactivation function in the pathogenesis of these diseases. Tackling these gaps of knowledge could reveal novel possible neuro-pathobiological mechanisms and present crucial information for the design of effective treatment modalities for devastating CNS diseases.


Asunto(s)
Síndrome de Angelman/metabolismo , Encéfalo/metabolismo , Ubiquitina-Proteína Ligasas/fisiología , Síndrome de Angelman/genética , Síndrome de Angelman/patología , Animales , Encéfalo/patología , Trastornos Generalizados del Desarrollo Infantil/genética , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Trastornos Generalizados del Desarrollo Infantil/patología , Humanos , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/patología , Unión Proteica/fisiología , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Síndrome de Rett/patología
18.
J Biol Chem ; 284(36): 24443-52, 2009 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-19561358

RESUMEN

In this study, we report that the purified wild-type FANCI (Fanconi anemia complementation group I) protein directly binds to a variety of DNA substrates. The DNA binding domain roughly encompasses residues 200-1000, as suggested by the truncation study. When co-expressed in insect cells, a small fraction of FANCI forms a stable complex with FANCD2 (Fanconi anemia complementation group D2). Intriguingly, the purified FANCI-FANCD2 complex preferentially binds to the branched DNA structures when compared with either FANCI or FANCD2 alone. Co-immunoprecipitation with purified proteins indicates that FANCI interacts with FANCD2 through its C-terminal amino acid 1001-1328 fragment. Although the C terminus of FANCI is dispensable for direct DNA binding, it seems to be involved in the regulation of DNA binding activity. This notion is further enhanced by two C-terminal point mutations, R1285Q and D1301A, which showed differentiated DNA binding activity. We also demonstrate that FANCI forms discrete nuclear foci in HeLa cells in the absence or presence of exogenous DNA damage. The FANCI foci are colocalized perfectly with FANCD2 and partially with proliferating cell nuclear antigen irrespective of mitomycin C treatment. An increased number of FANCI foci form and become resistant to Triton X extraction in response to mitomycin C treatment. Our data suggest that the FANCI-FANCD2 complex may participate in repair of damaged replication forks through its preferential recognition of branched structures.


Asunto(s)
Reparación del ADN/fisiología , ADN/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , Sustitución de Aminoácidos , Animales , ADN/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/química , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteínas del Grupo de Complementación de la Anemia de Fanconi/química , Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Expresión Génica , Células HeLa , Humanos , Mutación Puntual , Unión Proteica/fisiología , Estructura Terciaria de Proteína/fisiología , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Spodoptera
19.
Epilepsia ; 48(1): 175-81, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17241225

RESUMEN

PURPOSE: To determine if posthypoxia treatment with erythropoietin (EPO) has protective effects against subsequent susceptibility to seizure related neuronal injury in rat pups subjected to acute hypoxia at P10. METHODS: Four groups of rats were manipulated at P10, as described below, then all received kainic acid (KA) (10 mg/kg i.p.) at P29: Hypoxia-NS-KA group (n = 11): subjected to acute hypoxia (down to 4% O2), and then immediately received saline i.p. Hypoxia-EPO-KA group (n = 10): subjected to acute hypoxia and then immediately received EPO (1,000 U/Kg i.p.). Normoxia-NS-KA group (n = 11): sham manipulated and injected with saline. Normoxia-EPO-KA group (n = 10): sham manipulated then immediately injected with EPO (1000 U/Kg i.p.). After receiving KA at P29, all rats were monitored using videotape techniques, and were sacrificed at P31. TUNEL and Hoechst stains to assess for apoptosis, and regular histology for hippocampal cell counts were performed. RESULTS: Administration of the single dose of erythropoietin directly after an acute hypoxic event at P10 resulted at P29 in increased latency to forelimb clonus seizures, reduced duration of these seizures, protection against hippocampal cell loss, and decreased hippocampal apoptosis in the Hypoxia-EPO-KA group as compared to the Hypoxia-NS-KA group. CONCLUSION: These data support the presence of favorable protective effects of erythropoietin against the long-term consequences of acute hypoxia in the developing brain and raise the possibility of its investigation as a potential neuroprotective agent after human neonatal hypoxic encephalopathy.


Asunto(s)
Eritropoyetina/farmacología , Hipoxia/metabolismo , Convulsiones/inducido químicamente , Convulsiones/prevención & control , Animales , Animales Recién Nacidos , Recuento de Células , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/metabolismo , Relación Dosis-Respuesta a Droga , Eritropoyetina/administración & dosificación , Agonistas de Aminoácidos Excitadores/farmacología , Hipocampo/patología , Humanos , Hipoxia/patología , Hipoxia Encefálica/prevención & control , Ácido Kaínico/farmacología , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Ratas , Ratas Sprague-Dawley , Convulsiones/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA