Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Elife ; 122023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-38126351

RESUMEN

N6-methyladenine (6mA) DNA modification has recently been described in metazoans, including in Drosophila, for which the erasure of this epigenetic mark has been ascribed to the ten-eleven translocation (TET) enzyme. Here, we re-evaluated 6mA presence and TET impact on the Drosophila genome. Using axenic or conventional breeding conditions, we found traces of 6mA by LC-MS/MS and no significant increase in 6mA levels in the absence of TET, suggesting that this modification is present at very low levels in the Drosophila genome but not regulated by TET. Consistent with this latter hypothesis, further molecular and genetic analyses showed that TET does not demethylate 6mA but acts essentially in an enzymatic-independent manner. Our results call for further caution concerning the role and regulation of 6mA DNA modification in metazoans and underline the importance of TET non-enzymatic activity for fly development.


Asunto(s)
Adenina , Metilación de ADN , Proteínas de Drosophila , Drosophila , Animales , Cromatografía Liquida , ADN/genética , Drosophila/genética , Espectrometría de Masas en Tándem
2.
Sci Rep ; 13(1): 20832, 2023 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-38012215

RESUMEN

The COVID-19 pandemic demonstrated the need for rapid molecular diagnostics. Vaccination programs can provide protection and facilitate the opening of society, but newly emergent and existing viral variants capable of evading the immune system endanger their efficacy. Effective surveillance for Variants of Concern (VOC) is therefore important. Rapid and specific molecular diagnostics can provide speed and coverage advantages compared to genomic sequencing alone, benefitting the public health response and facilitating VOC containment. Here we expand the recently developed SARS-CoV-2 CRISPR-Cas detection technology (SHERLOCK) to provide rapid and sensitive discrimination of SARS-CoV-2 VOCs that can be used at point of care, implemented in the pipelines of small or large testing facilities, and even determine the proportion of VOCs in pooled population-level wastewater samples. This technology complements sequencing efforts to allow facile and rapid identification of individuals infected with VOCs to help break infection chains. We show the optimisation of our VarLOCK assays (Variant-specific SHERLOCK) for multiple specific mutations in the S gene of SARS-CoV-2 and validation with samples from the Cardiff University Testing Service. We also show the applicability of VarLOCK to national wastewater surveillance of SARS-CoV-2 variants and the rapid adaptability of the technique for new and emerging VOCs.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , SARS-CoV-2/genética , COVID-19/diagnóstico , COVID-19/epidemiología , Aguas Residuales , Pandemias , Monitoreo Epidemiológico Basado en Aguas Residuales , Pruebas en el Punto de Atención
3.
Brain Commun ; 5(2): fcad046, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36970044

RESUMEN

In humans and other primates, blood platelets contain high concentrations of brain-derived neurotrophic factor due to the expression of the BDNF gene in megakaryocytes. By contrast, mice, typically used to investigate the impact of CNS lesions, have no demonstrable levels of brain-derived neurotrophic factor in platelets, and their megakaryocytes do not transcribe significant levels of the Bdnf gene. Here, we explore potential contributions of platelet brain-derived neurotrophic factor with two well-established CNS lesion models, using 'humanized' mice engineered to express the Bdnf gene under the control of a megakaryocyte-specific promoter. Retinal explants prepared from mice containing brain-derived neurotrophic factor in platelets were labelled using DiOlistics and the dendritic integrity of retinal ganglion cells assessed after 3 days by Sholl analysis. The results were compared with retinas of wild-type animals and with wild-type explants supplemented with saturating concentrations of brain-derived neurotrophic factor or the tropomyosin kinase B antibody agonist, ZEB85. An optic nerve crush was also performed, and the dendrites of retinal ganglion cells similarly assessed 7-day post-injury, comparing the results of mice containing brain-derived neurotrophic factor in platelets with wild-type animals. In mice engineered to contain brain-derived neurotrophic factor in platelets, the mean serum brain-derived neurotrophic factor levels were 25.74 ± 11.36 ng/mL for homozygous and 17.02 ± 6.44 ng/mL for heterozygous mice, close to those determined in primates. Retinal explants from these animals showed robust preservation of dendrite complexity, similar to that seen with wild-type explants incubated with medium supplemented with brain-derived neurotrophic factor or the tropomyosin receptor kinase B antibody agonist, ZEB85. The Sholl areas under curve were 1811 ± 258, 1776 ± 435 and 1763 ± 256 versus 1406 ± 315 in the wild-type control group (P ≤ 0.001). Retinal ganglion cell survival based on cell counts was similar in all four groups, showing ∼15% loss. A robust neuroprotective effect was also observed following optic nerve crush when assessing the dendrites of the retinal ganglion cells in the transgenic mouse, with Sholl area under the curve significantly higher compared to wild-type (2667 ± 690 and 1921 ± 392, P = 0.026), with no significant difference in the contralateral eye controls. Repeat experiments found no difference in cell survival, with both showing ∼50% loss. These results indicate that platelet brain-derived neurotrophic factor has a strong neuroprotective effect on the dendrite complexity of retinal ganglion cells in both an ex vivo and in vivo model, suggesting that platelet brain-derived neurotrophic factor is likely to be a significant neuroprotective factor in primates.

4.
Sci Adv ; 8(36): eabm2427, 2022 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-36070377

RESUMEN

TET (ten-eleven translocation) enzymes catalyze the oxidation of 5-methylcytosine bases in DNA, thus driving active and passive DNA demethylation. Here, we report that the catalytic domain of mammalian TET enzymes favor CGs embedded within basic helix-loop-helix and basic leucine zipper domain transcription factor-binding sites, with up to 250-fold preference in vitro. Crystal structures and molecular dynamics calculations show that sequence preference is caused by intrasubstrate interactions and CG flanking sequence indirectly affecting enzyme conformation. TET sequence preferences are physiologically relevant as they explain the rates of DNA demethylation in TET-rescue experiments in culture and in vivo within the zygote and germ line. Most and least favorable TET motifs represent DNA sites that are bound by methylation-sensitive immediate-early transcription factors and octamer-binding transcription factor 4 (OCT4), respectively, illuminating TET function in transcriptional responses and pluripotency support.


Asunto(s)
5-Metilcitosina , Dioxigenasas , 5-Metilcitosina/metabolismo , Animales , Dominio Catalítico , Fenómenos Fisiológicos Celulares , ADN , Dioxigenasas/genética , Dioxigenasas/metabolismo , Mamíferos/genética
5.
Transl Psychiatry ; 11(1): 62, 2021 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-33462179

RESUMEN

Brain-derived neurotrophic factor (BDNF) plays crucial roles in brain function. Numerous studies report alterations in BDNF levels in human serum in various neurological conditions, including mood disorders such as depression. However, little is known about BDNF levels in the blood during pregnancy. We asked whether maternal depression and/or anxiety during pregnancy were associated with altered serum BDNF levels in mothers (n = 251) and their new-born infants (n = 212). As prenatal exposure to maternal mood disorders significantly increases the risk of neurological conditions in later life, we also examined the possibility of placental BDNF transfer by developing a new mouse model. We found no association between maternal symptoms of depression and either maternal or infant cord blood serum BDNF. However, maternal symptoms of anxiety correlated with significantly raised maternal serum BDNF exclusively in mothers of boys (r = 0.281; P = 0.005; n = 99). Serum BDNF was significantly lower in male infants than female infants but neither correlated with maternal anxiety symptoms. Consistent with this observation, we found no evidence for BDNF transfer across the placenta. We conclude that the placenta protects the developing fetus from maternal changes in serum BDNF that could otherwise have adverse consequences for fetal development.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Placenta , Ansiedad , Femenino , Sangre Fetal , Humanos , Masculino , Embarazo , Suero
6.
eNeuro ; 7(1)2020.
Artículo en Inglés | MEDLINE | ID: mdl-31882533

RESUMEN

While BDNF is receiving considerable attention for its role in synaptic plasticity and in nervous system dysfunction, identifying brain circuits involving BDNF-expressing neurons has been challenging. BDNF levels are very low in most brain areas, except for the large mossy fiber terminals in the hippocampus where BDNF accumulates at readily detectable levels. This report describes the generation of a mouse line allowing the detection of single brain cells synthesizing BDNF. A bicistronic construct encoding BDNF tagged with a P2A sequence preceding GFP allows the translation of BDNF and GFP as separate proteins. Following its validation with transfected cells, this construct was used to replace the endogenous Bdnf gene. Viable and fertile homozygote animals were generated, with the GFP signal marking neuronal cell bodies translating the Bdnf mRNA. Importantly, the distribution of immunoreactive BDNF remained unchanged, as exemplified by its accumulation in mossy fiber terminals in the transgenic animals. GFP-labeled neurons could be readily visualized in distinct layers in the cerebral cortex where BDNF has been difficult to detect with currently available reagents. In the hippocampal formation, quantification of the GFP signal revealed that <10% of the neurons do not translate the Bdnf mRNA at detectable levels, with the highest proportion of strongly labeled neurons found in CA3.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Hipocampo , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/metabolismo , Ratones , Plasticidad Neuronal , Neuronas/metabolismo
8.
J Neurosci ; 38(42): 9105-9121, 2018 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-30143575

RESUMEN

Specification of dorsoventral regional identity in progenitors of the developing telencephalon is a first pivotal step in the development of the cerebral cortex and basal ganglia. Previously, we demonstrated that the two zinc finger doublesex and mab-3 related (Dmrt) genes, Dmrt5 (Dmrta2) and Dmrt3, which are coexpressed in high caudomedial to low rostrolateral gradients in the cerebral cortical primordium, are separately needed for normal formation of the cortical hem, hippocampus, and caudomedial neocortex. We have now addressed the role of Dmrt3 and Dmrt5 in controlling dorsoventral division of the telencephalon in mice of either sex by comparing the phenotypes of single knock-out (KO) with double KO embryos and by misexpressing Dmrt5 in the ventral telencephalon. We find that DMRT3 and DMRT5 act as critical regulators of progenitor cell dorsoventral identity by repressing ventralizing regulators. Early ventral fate transcriptional regulators expressed in the dorsal lateral ganglionic eminence, such as Gsx2, are upregulated in the dorsal telencephalon of Dmrt3;Dmrt5 double KO embryos and downregulated when ventral telencephalic progenitors express ectopic Dmrt5 Conditional overexpression of Dmrt5 throughout the telencephalon produces gene expression and structural defects that are highly consistent with reduced GSX2 activity. Further, Emx2;Dmrt5 double KO embryos show a phenotype similar to Dmrt3;Dmrt5 double KO embryos, and both DMRT3, DMRT5 and the homeobox transcription factor EMX2 bind to a ventral telencephalon-specific enhancer in the Gsx2 locus. Together, our findings uncover cooperative functions of DMRT3, DMRT5, and EMX2 in dividing dorsal from ventral in the telencephalon.SIGNIFICANCE STATEMENT We identified the DMRT3 and DMRT5 zinc finger transcription factors as novel regulators of dorsoventral patterning in the telencephalon. Our data indicate that they have overlapping functions and compensate for one another. The double, but not the single, knock-out produces a dorsal telencephalon that is ventralized, and olfactory bulb tissue takes over most remaining cortex. Conversely, overexpressing Dmrt5 throughout the telencephalon causes expanded expression of dorsal gene determinants and smaller olfactory bulbs. Furthermore, we show that the homeobox transcription factor EMX2 that is coexpressed with DMRT3 and DMRT5 in cortical progenitors cooperates with them to maintain dorsoventral patterning in the telencephalon. Our study suggests that DMRT3/5 function with EMX2 in positioning the pallial-subpallial boundary by antagonizing the ventral homeobox transcription factor GSX2.


Asunto(s)
Proteínas de Homeodominio/fisiología , Células-Madre Neurales/fisiología , Neuronas/fisiología , Telencéfalo/embriología , Factores de Transcripción/fisiología , Animales , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Telencéfalo/metabolismo , Factores de Transcripción/genética
9.
Proc Natl Acad Sci U S A ; 115(30): E7023-E7032, 2018 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-29987039

RESUMEN

The diverse physiological roles of the neurotrophin family have long prompted exploration of their potential as therapeutic agents for nerve injury and neurodegenerative diseases. To date, clinical trials of one family member, brain-derived neurotrophic factor (BDNF), have disappointingly failed to meet desired endpoints. Contributing to these failures is the fact that BDNF is pharmaceutically a nonideal biologic drug candidate. It is a highly charged, yet is a net hydrophobic molecule with a low molecular weight that confers a short t1/2 in man. To circumvent these shortcomings of BDNF as a drug candidate, we have employed a function-based cellular screening assay to select activating antibodies of the BDNF receptor TrkB from a combinatorial human short-chain variable fragment antibody library. We report here the successful selection of several potent TrkB agonist antibodies and detailed biochemical and physiological characterization of one such antibody, ZEB85. By using a human TrkB reporter cell line and BDNF-responsive GABAergic neurons derived from human ES cells, we demonstrate that ZEB85 is a full agonist of TrkB, comparable in potency to BDNF toward human neurons in activation of TrkB phosphorylation, canonical signal transduction, and mRNA transcriptional regulation.


Asunto(s)
Comunicación Autocrina , Neuronas GABAérgicas/metabolismo , Biblioteca de Genes , Glicoproteínas de Membrana/agonistas , Receptor trkB/agonistas , Transducción de Señal/efectos de los fármacos , Anticuerpos de Cadena Única , Transcripción Genética/efectos de los fármacos , Línea Celular , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Fosforilación/efectos de los fármacos , Receptor trkB/genética , Receptor trkB/metabolismo , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/farmacología
10.
Cereb Cortex ; 28(2): 493-509, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28031177

RESUMEN

Mice that are constitutively null for the zinc finger doublesex and mab-3 related (Dmrt) gene, Dmrt5/Dmrta2, show a variety of patterning abnormalities in the cerebral cortex, including the loss of the cortical hem, a powerful cortical signaling center. In conditional Dmrt5 gain of function and loss of function mouse models, we generated bidirectional changes in the neocortical area map without affecting the hem. Analysis indicated that DMRT5, independent of the hem, directs the rostral-to-caudal pattern of the neocortical area map. Thus, DMRT5 joins a small number of transcription factors shown to control directly area size and position in the neocortex. Dmrt5 deletion after hem formation also reduced hippocampal size and shifted the position of the neocortical/paleocortical boundary. Dmrt3, like Dmrt5, is expressed in a gradient across the cortical primordium. Mice lacking Dmrt3 show cortical patterning defects akin to but milder than those in Dmrt5 mutants, perhaps in part because Dmrt5 expression increases in the absence of Dmrt3. DMRT5 upregulates Dmrt3 expression and negatively regulates its own expression, which may stabilize the level of DMRT5. Together, our findings indicate that finely tuned levels of DMRT5, together with DMRT3, regulate patterning of the cerebral cortex.


Asunto(s)
Desarrollo Embrionario/fisiología , Hipocampo/metabolismo , Neocórtex/metabolismo , Factores de Transcripción/biosíntesis , Animales , Hipocampo/embriología , Hipocampo/crecimiento & desarrollo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Neocórtex/embriología , Neocórtex/crecimiento & desarrollo , Neurogénesis/fisiología
11.
Proc Natl Acad Sci U S A ; 114(28): E5599-E5607, 2017 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-28655839

RESUMEN

The mechanisms that determine whether a neural progenitor cell (NPC) reenters the cell cycle or exits and differentiates are pivotal for generating cells in the correct numbers and diverse types, and thus dictate proper brain development. Combining gain-of-function and loss-of-function approaches in an embryonic stem cell-derived cortical differentiation model, we report that doublesex- and mab-3-related transcription factor a2 (Dmrta2, also known as Dmrt5) plays an important role in maintaining NPCs in the cell cycle. Temporally controlled expression of transgenic Dmrta2 in NPCs suppresses differentiation without affecting their neurogenic competence. In contrast, Dmrta2 knockout accelerates the cell cycle exit and differentiation into postmitotic neurons of NPCs derived from embryonic stem cells and in Emx1-cre conditional mutant mice. Dmrta2 function is linked to the regulation of Hes1 and other proneural genes, as demonstrated by genome-wide RNA-seq and direct binding of Dmrta2 to the Hes1 genomic locus. Moreover, transient Hes1 expression rescues precocious neurogenesis in Dmrta2 knockout NPCs. Our study thus establishes a link between Dmrta2 modulation of Hes1 expression and the maintenance of NPCs during cortical development.


Asunto(s)
Células-Madre Neurales/citología , Factor de Transcripción HES-1/genética , Factor de Transcripción HES-1/fisiología , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Animales , Ciclo Celular , Diferenciación Celular , Proliferación Celular , Células Madre Embrionarias/citología , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Noqueados , Neurogénesis , Neuronas/citología , Fenotipo , Transgenes
12.
Sci Rep ; 6: 32488, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27580818

RESUMEN

Cell type-specific surface markers offer a powerful tool for purifying defined cell types for restorative therapies and drug screenings. Midbrain dopaminergic neurons (mesDA) are the nerve cells preferentially lost in the brains of Parkinson's disease patients. Clinical trials of transplantation of fetal neural precursors suggest that cell therapy may offer a cure for this devastating neurological disease. Many lines of preclinical studies demonstrate that neural progenitors committed to dopaminergic fate survive and integrate better than postmitotic DA neurons. We show that the folate-receptor 1 (FolR1), a GPI-anchored cell surface molecule, specifically marks mesDA neural progenitors and immature mesDA neurons. FolR1 expression superimposes with Lmx1a, a bona-fide mesDA lineage marker, during the active phase of mesDA neurogenesis from E9.5 to E14.5 during mouse development, as well as in ESC-derived mesDA lineage. FolR1(+) neural progenitors can be isolated by FACS or magnetic sorting (MAC) which give rise to dopamine neurons expressing TH and Pitx3, whilst FolR1 negative cells generate non-dopaminergic neurons and glia cells. This study identifies FolR1 as a new cell surface marker selectively expressed in mesDA progenitors in vivo and in vitro and that can be used to enrich in vitro differentiated TH neurons.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Receptor 1 de Folato/genética , Mesencéfalo/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Células-Madre Neurales/metabolismo , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Línea Celular , Separación Celular/métodos , Dopamina/metabolismo , Neuronas Dopaminérgicas/citología , Embrión de Mamíferos , Receptor 1 de Folato/metabolismo , Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Mesencéfalo/citología , Ratones , Células Madre Embrionarias de Ratones/citología , Células-Madre Neurales/citología , Neurogénesis/genética , Neuroglía/citología , Neuroglía/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Tirosina 3-Monooxigenasa/genética , Tirosina 3-Monooxigenasa/metabolismo
13.
Proc Natl Acad Sci U S A ; 108(22): 9131-6, 2011 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-21576465

RESUMEN

Understanding the control of cell-fate choices during embryonic stem cell (ESC) differentiation is crucial for harnessing strategies for efficient production of desired cell types for pharmaceutical drug screening and cell transplantation. Here we report the identification of the zinc finger-like doublesex and mab-3-related transcription factor 5 (Dmrt5) as a marker for mammalian ventral-medial mesencephalic neuroepithelium that give rise to dopamine neurons. Gain- and loss-of-function studies in ESC demonstrate that Dmrt5 is critically involved in the specification of ventral-medial neural progenitor cell fate and the subsequent generation of dopamine neurons expressing essential midbrain characteristics. Genome-wide analysis of Dmrt5-mediated transcriptome changes and expression profiling of ventral-medial and ventral-lateral mesencephalic neuroepithelium revealed suppressive and inductive regulatory roles for Dmrt5 in the transcription program associated with the ventral-medial neural progenitor fates. Together, these data identify Dmrt5 as an important player in ventral mesencephalic neural fate specification.


Asunto(s)
Dopamina/metabolismo , Mesencéfalo/metabolismo , Células Madre Pluripotentes/citología , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Animales , Encéfalo/metabolismo , Linaje de la Célula , Células , Embrión de Pollo , Células Madre Embrionarias/citología , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Ratones , Neuronas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Células Madre/citología
14.
Proc Natl Acad Sci U S A ; 104(8): 2709-14, 2007 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-17296936

RESUMEN

Mutations in the human methyl-CpG-binding protein gene MECP2 cause the neurological disorder Rett syndrome and some cases of X-linked mental retardation (XLMR). We report that MeCP2 interacts with ATRX, a SWI2/SNF2 DNA helicase/ATPase that is mutated in ATRX syndrome (alpha-thalassemia/mental retardation, X-linked). MeCP2 can recruit the helicase domain of ATRX to heterochromatic foci in living mouse cells in a DNA methylation-dependent manner. Also, ATRX localization is disrupted in neurons of Mecp2-null mice. Point mutations within the methylated DNA-binding domain of MeCP2 that cause Rett syndrome or X-linked mental retardation inhibit its interaction with ATRX in vitro and its localization in vivo without affecting methyl-CpG binding. We propose that disruption of the MeCP2-ATRX interaction leads to pathological changes that contribute to mental retardation.


Asunto(s)
ADN Helicasas/metabolismo , Discapacidad Intelectual/genética , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Mutación/genética , Proteínas Nucleares/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Células Cultivadas , ADN/metabolismo , ADN Helicasas/química , Metilación de ADN , Humanos , Proteína 2 de Unión a Metil-CpG/deficiencia , Ratones , Proteínas Nucleares/química , Unión Proteica , Transporte de Proteínas , Técnicas del Sistema de Dos Híbridos , Proteína Nuclear Ligada al Cromosoma X
15.
EMBO J ; 25(22): 5317-28, 2006 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17066076

RESUMEN

In mammalian cells, DNA methylation is associated with heritable and stable gene repression, mediated in part by methyl-CpG-binding domain (MBD) proteins that recruit corepressors to modify chromatin. MBD1 protein, a member of the MBD family, forms a complex with SETDB1 histone methylase to silence transcription at target promoters by methylation of lysine 9 of histone H3. How MBD1-mediated transcriptional repression is regulated is currently unknown. Here we show that MBD1 is a target for sumoylation by PIAS1 (Protein Inhibitors of Activated STAT 1) and PIAS3 E3 SUMO (small ubiquitin-like modifier)-ligases, at two conserved lysine residues within the C-terminus of MBD1. Although sumoylated MBD1 binds to methylated DNA, it does not incorporate into a complex with SETDB1 and does not efficiently repress transcription of a target gene, p53BP2, in HeLa cells. Our data suggest that transcriptional silencing by MBD1 is regulated by a PIAS-mediated conjugation of SUMO1, which antagonizes the formation of a repressive complex with SETDB1.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Chaperonas Moleculares/fisiología , Proteínas Inhibidoras de STAT Activados/fisiología , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/fisiología , Factores de Transcripción/fisiología , Transcripción Genética , Secuencia de Aminoácidos , Proteínas Reguladoras de la Apoptosis , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas Portadoras/fisiología , Cromatina/genética , Cromatina/fisiología , Secuencia Conservada , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo , Silenciador del Gen , Células HeLa , N-Metiltransferasa de Histona-Lisina , Histonas/metabolismo , Humanos , Chaperonas Moleculares/genética , Datos de Secuencia Molecular , Unión Proteica , Proteínas Inhibidoras de STAT Activados/genética , Proteína Metiltransferasas/metabolismo , Proteína SUMO-1 , Homología de Secuencia de Aminoácido , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/genética , Factores de Transcripción/metabolismo
17.
Biochem Biophys Res Commun ; 324(1): 348-54, 2004 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-15465025

RESUMEN

Transcription activity of chromatin is associated with histone acetylation which is regulated by recruitment of histone acetyltransferases and deacetylases (HDAC) to specific chromatin regions. We have tested how expression of a transfected or stably introduced gene correlates with histone acetylation. Our results demonstrate that expression of transiently transfected green fluorescence protein (GFP) genes is significantly enhanced by HDAC inhibitors. Although HDAC treatment did not induce noticeable changes in the chromatin structure of genomic DNA, chromatin immunoprecipitation showed that more transiently transfected DNA is assembled into chromatin containing acetylated histones in HDAC inhibitor treated cells when compared to untreated cells. For stably integrated GFP, the expression response to HDAC inhibitors varies between independent stable cell lines. However, there was no difference in histone acetylation associated with the integrated transgene between HDAC inhibitor responsive and non-responsive cells. Furthermore, the overall enhancement of transgene expression by HDAC inhibitors was not as pronounced as in transiently transfected cells.


Asunto(s)
ADN/metabolismo , Regulación de la Expresión Génica , Inhibidores de Histona Desacetilasas , Transfección , Acetilación , Animales , Células CHO , Cricetinae , ADN/química , Inhibidores Enzimáticos/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Histona Desacetilasas/metabolismo , Ácidos Hidroxámicos/metabolismo , Conformación de Ácido Nucleico , Transcripción Genética , Transgenes
18.
J Control Release ; 99(3): 435-44, 2004 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-15451601

RESUMEN

The protein transduction domain (PTD) of the HIV-1 Tat protein can facilitate the cellular and nuclear uptake of macromolecular particles. Here, we demonstrate that incorporation without covalent linkage of a 17-amino acid PTD peptide into gene delivery lipoplexes improves gene transfer. Tat/Liposome/DNA (TLD) transfection, as evaluated by Fluorescence Activated Cell Scan analysis of a Green Fluorescence Protein expression plasmid, enabled transfection of highly recalcitrant primary cells in the form of air/liquid interface cultures of sheep tracheal epithelium. Treatment with chloroquine increased, and incubation at low temperature decreased, TLD transfection, suggesting that the endocytosis uptake pathway is involved.


Asunto(s)
Productos del Gen tat/farmacocinética , VIH-1/fisiología , Liposomas/farmacocinética , Transfección/métodos , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/farmacocinética , Línea Celular Tumoral , Cloroquina/farmacología , ADN/genética , ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Desoxirribonucleasa I/efectos de los fármacos , Desoxirribonucleasa I/fisiología , Evaluación Preclínica de Medicamentos/métodos , Ensayo de Cambio de Movilidad Electroforética/métodos , Endocitosis/efectos de los fármacos , Endocitosis/fisiología , Ácidos Grasos Monoinsaturados/química , Ácidos Grasos Monoinsaturados/farmacocinética , Citometría de Flujo/métodos , Predicción , Productos del Gen tat/genética , Terapia Genética/métodos , Vectores Genéticos/genética , Vectores Genéticos/farmacocinética , VIH-1/genética , Humanos , Liposomas/química , Sustancias Macromoleculares/química , Tamaño de la Partícula , Fosfatidiletanolaminas/farmacocinética , Plásmidos/química , Plásmidos/farmacocinética , Compuestos de Amonio Cuaternario/química , Compuestos de Amonio Cuaternario/farmacocinética , Análisis de Secuencia de Proteína/métodos , Ovinos , Tráquea/efectos de los fármacos , Tráquea/patología , Tráquea/fisiología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
19.
J Biol Chem ; 278(6): 4035-40, 2003 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-12427740

RESUMEN

DNA methylation plays an important role in mammalian development and correlates with chromatin-associated gene silencing. The recruitment of MeCP2 to methylated CpG dinucleotides represents a major mechanism by which DNA methylation can repress transcription. MeCP2 silences gene expression partly by recruiting histone deacetylase (HDAC) activity, resulting in chromatin remodeling. Here, we show that MeCP2 associates with histone methyltransferase activity in vivo and that this activity is directed against Lys(9) of histone H3. Two characterized repression domains of MeCP2 are involved in tethering the histone methyltransferase to MeCP2. We asked if MeCP2 can deliver Lys(9) H3 methylation to the H19 gene, whose activity it represses. We show that the presence of MeCP2 on nucleosomes within the repressor region of the H19 gene (the differentially methylated domain) coincides with an increase in H3 Lys(9) methylation. Our data provide evidence that MeCP2 reinforces a repressive chromatin state by acting as a bridge between two global epigenetic modifications, DNA methylation and histone methylation.


Asunto(s)
Proteínas Cromosómicas no Histona , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Histonas/metabolismo , Proteínas Represoras , Animales , Línea Celular , Histona Desacetilasas/metabolismo , Humanos , Proteína 2 de Unión a Metil-CpG , Metilación , Ratones , Pruebas de Precipitina , Proteínas Recombinantes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Hereditas ; 139(2): 90-5, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-15061809

RESUMEN

Here we describe our comparative studies on two types of X chromosomes, namely X(M) and X(SM,) of the mandarin vole (Microtus mandarinus). By chromosome G- and C-banding analysis, we have found that two different types of X chromosomes exist in mandarin voles. The two types of X chromosomes present two different G- and C-banding patterns: the X(M) chromosome is a longer metacentric X chromosome which is C-band negative; and the X(SM) is a shorter submetacentric X chromosome which has one C-band at the centromere and another one at the middle part of the short arm. The X(SM) has 6 G-bands including one on the kinetochore, one in the middle of the short arm, and four on the long arm. The X(M) has 7 G-bands including one on the kinetochore, two on the short arm, and four on the long arm. We have further found that female voles can be grouped into three types based on the composition of the X chromosome but the male voles have only one type. The three female groups are: (1) female voles (X(M)X(SM)), in which the two X chromosomes are different, the longer one is metacentric and the shorter is submetacentric; (2) female vole (X(SM)X(SM)), in which the two X chromosomes are both submetacentric; (3) female vole (X(M)O), in which there is only one X chromosome that is metacentric. Surprisingly, we have never found female voles with X(M)X(M), females with X(SM)O or males with X(M)Y. We hypothesize that the X(SM) chromosome is derived from the X(M) through its breakage and re-joining. The paper also discusses the formation of X(M)O females.


Asunto(s)
Arvicolinae/genética , Variación Genética , Cromosoma X , Animales , Bandeo Cromosómico , Femenino , Cariotipificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA