Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Cell Rep ; 42(4): 112353, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37043353

RESUMEN

Stem cell therapy shows promise for multiple disorders; however, the molecular crosstalk between grafted cells and host tissue is largely unknown. Here, we take a step toward addressing this question. Using translating ribosome affinity purification (TRAP) with sequencing tools, we simultaneously decode the transcriptomes of graft and host for human neural stem cells (hNSCs) transplanted into the stroke-injured rat brain. Employing pathway analysis tools, we investigate the interactions between the two transcriptomes to predict molecular pathways linking host and graft genes; as proof of concept, we predict host-secreted factors that signal to the graft and the downstream molecular cascades they trigger in the graft. We identify a potential host-graft crosstalk pathway where BMP6 from the stroke-injured brain induces graft secretion of noggin, a known brain repair factor. Decoding the molecular interplay between graft and host is a critical step toward deciphering the molecular mechanisms of stem cell action.


Asunto(s)
Células-Madre Neurales , Accidente Cerebrovascular , Ratas , Animales , Humanos , Encéfalo , Accidente Cerebrovascular/terapia , Trasplante de Células Madre , Diferenciación Celular
3.
Stem Cells Transl Med ; 6(10): 1917-1929, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28834391

RESUMEN

Cervical spinal cord injury (SCI) remains an important research focus for regenerative medicine given the potential for severe functional deficits and the current lack of treatment options to augment neurological recovery. We recently reported the preclinical safety data of a human embryonic cell-derived oligodendrocyte progenitor cell (OPC) therapy that supported initiation of a phase I clinical trial for patients with sensorimotor complete thoracic SCI. To support the clinical use of this OPC therapy for cervical injuries, we conducted preclinical efficacy and safety testing of the OPCs in a nude rat model of cervical SCI. Using the automated TreadScan system to track motor behavioral recovery, we found that OPCs significantly improved locomotor performance when administered directly into the cervical spinal cord 1 week after injury, and that this functional improvement was associated with reduced parenchymal cavitation and increased sparing of myelinated axons within the injury site. Based on large scale biodistribution and toxicology studies, we show that OPC migration is limited to the spinal cord and brainstem and did not cause any adverse clinical observations, toxicities, allodynia, or tumors. In combination with previously published efficacy and safety data, the results presented here supported initiation of a phase I/IIa clinical trial in the U.S. for patients with sensorimotor complete cervical SCI. Stem Cells Translational Medicine 2017;6:1917-1929.


Asunto(s)
Células Madre Embrionarias Humanas/citología , Células-Madre Neurales/trasplante , Oligodendroglía/trasplante , Traumatismos de la Médula Espinal/terapia , Trasplante de Células Madre/efectos adversos , Animales , Movimiento Celular , Vértebras Cervicales/lesiones , Femenino , Humanos , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Oligodendroglía/citología , Oligodendroglía/fisiología , Ratas , Trasplante de Células Madre/métodos
4.
Regen Med ; 10(8): 939-58, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26345388

RESUMEN

AIM: To characterize the preclinical safety profile of a human embryonic stem cell-derived oligodendrocyte progenitor cell therapy product (AST-OPC1) in support of its use as a treatment for spinal cord injury (SCI). MATERIALS & METHODS: The phenotype and functional capacity of AST-OPC1 was characterized in vitro and in vivo. Safety and toxicology of AST-OPC1 administration was assessed in rodent models of thoracic SCI. RESULTS: These results identify AST-OPC1 as an early-stage oligodendrocyte progenitor population capable of promoting neurite outgrowth in vitro and myelination in vivo. AST-OPC1 administration did not cause any adverse clinical observations, toxicities, allodynia or tumors. CONCLUSION: These results supported initiation of a Phase I clinical trial in patients with sensorimotor complete thoracic SCI.


Asunto(s)
Células Madre Embrionarias Humanas , Oligodendroglía , Traumatismos de la Médula Espinal/terapia , Trasplante de Células Madre/métodos , Animales , Xenoinjertos , Células Madre Embrionarias Humanas/metabolismo , Células Madre Embrionarias Humanas/trasplante , Humanos , Ratones , Ratones Desnudos , Oligodendroglía/metabolismo , Oligodendroglía/trasplante , Traumatismos de la Médula Espinal/metabolismo , Trasplante de Células Madre/efectos adversos
5.
PLoS One ; 9(10): e109803, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25299045

RESUMEN

The obligate intracellular parasite, Toxoplasma gondii, disseminates through its host inside infected immune cells. We hypothesize that parasite nutrient requirements lead to manipulation of migratory properties of the immune cell. We demonstrate that 1) T. gondii relies on glutamine for optimal infection, replication and viability, and 2) T. gondii-infected bone marrow-derived dendritic cells (DCs) display both "hypermotility" and "enhanced migration" to an elevated glutamine gradient in vitro. We show that glutamine uptake by the sodium-dependent neutral amino acid transporter 2 (SNAT2) is required for this enhanced migration. SNAT2 transport of glutamine is also a significant factor in the induction of migration by the small cytokine stromal cell-derived factor-1 (SDF-1) in uninfected DCs. Blocking both SNAT2 and C-X-C chemokine receptor 4 (CXCR4; the unique receptor for SDF-1) blocks hypermotility and the enhanced migration in T. gondii-infected DCs. Changes in host cell protein expression following T. gondii infection may explain the altered migratory phenotype; we observed an increase of CD80 and unchanged protein level of CXCR4 in both T. gondii-infected and lipopolysaccharide (LPS)-stimulated DCs. However, unlike activated DCs, SNAT2 expression in the cytosol of infected cells was also unchanged. Thus, our results suggest an important role of glutamine transport via SNAT2 in immune cell migration and a possible interaction between SNAT2 and CXCR4, by which T. gondii manipulates host cell motility.


Asunto(s)
Sistemas de Transporte de Aminoácidos/genética , Células Dendríticas/parasitología , Glutamina/metabolismo , Receptores CXCR4/genética , Toxoplasma/genética , Sistema de Transporte de Aminoácidos A , Sistemas de Transporte de Aminoácidos/inmunología , Animales , Animales Recién Nacidos , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/parasitología , Células de la Médula Ósea/patología , Diferenciación Celular , Movimiento Celular/efectos de los fármacos , Quimiocina CXCL12/genética , Quimiocina CXCL12/inmunología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/patología , Fibroblastos/inmunología , Fibroblastos/parasitología , Fibroblastos/patología , Regulación de la Expresión Génica , Genes Reporteros , Glutamina/farmacología , Interacciones Huésped-Patógeno , Humanos , Lipopolisacáridos/farmacología , Luciferasas/genética , Luciferasas/metabolismo , Organismos Modificados Genéticamente , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Receptores CXCR4/inmunología , Transducción de Señal , Toxoplasma/inmunología , Toxoplasma/metabolismo
6.
Neuroendocrinology ; 100(2-3): 129-40, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25228100

RESUMEN

BACKGROUND/AIMS: Stress exacerbates neuron loss in many CNS injuries via the actions of adrenal glucocorticoid (GC) hormones. For some injuries, this GC endangerment of neurons is accompanied by greater immune cell activation in the CNS, a surprising outcome given the potent immunosuppressive properties of GCs. METHODS: To determine whether the effects of GCs on inflammation contribute to neuron death or result from it, we tested whether nonsteroidal anti-inflammatory drugs could protect neurons from GCs during kainic acid excitotoxicity in adrenalectomized male rats. We next measured GC effects on (1) chemokine production (CCL2 and CINC-1), (2) signals that suppress immune activation (CX3CL1, CD22, CD200, and TGF-ß), and (3) NF-κB activity. RESULTS: Concurrent treatment with minocycline, but not indomethacin, prevented GC endangerment. GCs did not substantially affect CCL2, CINC-1, or baseline NF-κB activity, but they did suppress CX3CL1, CX3CR1, and CD22 expression in the hippocampus - factors that normally restrain inflammatory responses. CONCLUSIONS: These findings demonstrate that cellular inflammation is not necessarily suppressed by GCs in the injured hippocampus; instead, GCs may worsen hippocampal neuron death, at least in part by increasing the neurotoxicity of CNS inflammation.


Asunto(s)
Agonistas de Aminoácidos Excitadores/toxicidad , Glucocorticoides/metabolismo , Hipocampo/efectos de los fármacos , Hipocampo/inmunología , Ácido Kaínico/toxicidad , Adrenalectomía , Animales , Antiinflamatorios no Esteroideos/farmacología , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Quimiocinas/metabolismo , Relación Dosis-Respuesta a Droga , Hipocampo/patología , Indometacina/farmacología , Masculino , Minociclina/farmacología , Neuroinmunomodulación/efectos de los fármacos , Neuroinmunomodulación/fisiología , Neuronas/efectos de los fármacos , Neuronas/inmunología , Neuronas/patología , Fármacos Neuroprotectores/farmacología , Ratas Sprague-Dawley
7.
Stem Cell Res Ther ; 5(2): 45, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24690461

RESUMEN

INTRODUCTION: Stroke is the second leading cause of death worldwide, claims six lives every 60 seconds, and is a leading cause of adult disability across the globe. Tissue plasminogen activator, the only United States Food and Drug Administration (FDA)-approved drug currently available, has a narrow therapeutic time window of less than 5 hours. In the past decade, cells derived from the human umbilical cord (HUC) have emerged as a potential therapeutic alternative for stroke; however, the most effective HUC-derived cell population remains unknown. METHODS: We compared three cell populations derived from the human umbilical cord: cord blood mononuclear cells (cbMNCs); cord blood mesenchymal stromal cells (cbMSCs), a subpopulation of cbMNCs; and cord matrix MSCs (cmMSCs). We characterized these cells in vitro with flow cytometry and assessed the cells' in vivo efficacy in a 2-hour transient middle cerebral artery occlusion (MCAo) rat model of stroke. cbMNCs, cbMSCs, and cmMSCs were each transplanted intraarterially at 24 hours after stroke. RESULTS: A reduction in neurologic deficit and infarct area was observed in all three cell groups; however, this reduction was significantly enhanced in the cbMNC group compared with the cmMSC group. At 2 weeks after stroke, human nuclei-positive cells were present in the ischemic hemispheres of immunocompetent stroke rats in all three cell groups. Significantly decreased expression of rat brain-derived neurotrophic factor mRNA was observed in the ischemic hemispheres of all three cell-treated and phosphate-buffered saline (PBS) group animals compared with sham animals, although the decrease was least in cbMNC-treated animals. Significantly decreased expression of rat interleukin (IL)-2 mRNA and IL-6 mRNA was seen only in the cbMSC group. Notably, more severe complications (death, eye inflammation) were observed in the cmMSC group compared with the cbMNC and cbMSC groups. CONCLUSIONS: All three tested cell types promoted recovery after stroke, but cbMNCs showed enhanced recovery and fewer complications compared with cmMSCs.


Asunto(s)
Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Infarto de la Arteria Cerebral Media/terapia , Leucocitos Mononucleares/trasplante , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Animales , Diferenciación Celular/fisiología , Modelos Animales de Enfermedad , Humanos , Infusiones Intraarteriales , Leucocitos Mononucleares/citología , Masculino , Distribución Aleatoria , Ratas , Ratas Wistar
8.
PLoS One ; 8(4): e61789, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23613937

RESUMEN

Research with experimental stroke models has identified a wide range of therapeutic proteins that can prevent the brain damage caused by this form of acute neurological injury. Despite this, we do not yet have safe and effective ways to deliver therapeutic proteins to the injured brain, and this remains a major obstacle for clinical translation. Current targeted strategies typically involve invasive neurosurgery, whereas systemic approaches produce the undesirable outcome of non-specific protein delivery to the entire brain, rather than solely to the injury site. As a potential way to address this, we developed a protein delivery system modeled after the endogenous immune cell response to brain injury. Using ex-vivo-engineered dendritic cells (DCs), we find that these cells can transiently home to brain injury in a rat model of stroke with both temporal and spatial selectivity. We present a standardized method to derive injury-responsive DCs from bone marrow and show that injury targeting is dependent on culture conditions that maintain an immature DC phenotype. Further, we find evidence that when loaded with therapeutic cargo, cultured DCs can suppress initial neuron death caused by an ischemic injury. These results demonstrate a non-invasive method to target ischemic brain injury and may ultimately provide a way to selectively deliver therapeutic compounds to the injured brain.


Asunto(s)
Encéfalo/metabolismo , Células Dendríticas , Proteínas/administración & dosificación , Proteínas/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Lesiones Encefálicas/tratamiento farmacológico , Modelos Animales de Enfermedad , Ratas
9.
Curr Pharm Des ; 18(25): 3685-93, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22571604

RESUMEN

Stem cell transplantation has emerged as a promising treatment strategy for stroke. The development of effective ways to monitor transplanted stem cells is essential to understand how stem cell transplantation enhances stroke recovery and ultimately will be an indispensable tool for advancing stem cell therapy to the clinic. In this review, we describe existing methods of tracking transplanted stem cells in vivo, including optical imaging, magnetic resonance imaging (MRI), and positron emission tomography (PET), with emphasis on the benefits and drawbacks of each imaging approach. Key considerations such as the potential impact of each tracking system on stem cell function, as well as its relative applicability to humans are discussed. Finally, we describe multi-modal imaging strategies as a more comprehensive method to track transplanted stem cells in the stroke-injured brain.


Asunto(s)
Trasplante de Células Madre , Células Madre/patología , Accidente Cerebrovascular/terapia , Humanos , Imagen por Resonancia Magnética , Tomografía de Emisión de Positrones , Accidente Cerebrovascular/patología
10.
Proc Natl Acad Sci U S A ; 109(14): 5475-80, 2012 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-22431614

RESUMEN

Stroke causes brain dysfunction and neuron death, and the lack of effective therapies heightens the need for new therapeutic targets. Here we identify prokineticin 2 (PK2) as a mediator for cerebral ischemic injury. PK2 is a bioactive peptide initially discovered as a regulator of gastrointestinal motility. Multiple biological roles for PK2 have been discovered, including circadian rhythms, angiogenesis, and neurogenesis. However, the role of PK2 in neuropathology is unknown. Using primary cortical cultures, we found that PK2 mRNA is up-regulated by several pathological stressors, including hypoxia, reactive oxygen species, and excitotoxic glutamate. Glutamate-induced PK2 expression is dependent on NMDA receptor activation and extracellular calcium. Enriched neuronal culture studies revealed that neurons are the principal source of glutamate-induced PK2. Using in vivo models of stroke, we found that PK2 mRNA is induced in the ischemic cortex and striatum. Central delivery of PK2 worsens infarct volume, whereas PK2 receptor antagonist decreases infarct volume and central inflammation while improving functional outcome. Direct central inhibition of PK2 using RNAi also reduces infarct volume. These findings indicate that PK2 can be activated by pathological stimuli such as hypoxia-ischemia and excitotoxic glutamate and identify PK2 as a deleterious mediator for cerebral ischemia.


Asunto(s)
Isquemia Encefálica/fisiopatología , Hormonas Gastrointestinales/fisiología , Neuropéptidos/fisiología , Animales , Hormonas Gastrointestinales/genética , Neuropéptidos/genética , ARN Mensajero/genética , Ratas , Regulación hacia Arriba
11.
Brain ; 134(Pt 6): 1777-89, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21616972

RESUMEN

Stem cell transplantation promises new hope for the treatment of stroke although significant questions remain about how the grafted cells elicit their effects. One hypothesis is that transplanted stem cells enhance endogenous repair mechanisms activated after cerebral ischaemia. Recognizing that bilateral reorganization of surviving circuits is associated with recovery after stroke, we investigated the ability of transplanted human neural progenitor cells to enhance this structural plasticity. Our results show the first evidence that human neural progenitor cell treatment can significantly increase dendritic plasticity in both the ipsi- and contralesional cortex and this coincides with stem cell-induced functional recovery. Moreover, stem cell-grafted rats demonstrated increased corticocortical, corticostriatal, corticothalamic and corticospinal axonal rewiring from the contralesional side; with the transcallosal and corticospinal axonal sprouting correlating with functional recovery. Furthermore, we demonstrate that axonal transport, which is critical for both proper axonal function and axonal sprouting, is inhibited by stroke and that this is rescued by the stem cell treatment, thus identifying another novel potential mechanism of action of transplanted cells. Finally, we established in vitro co-culture assays in which these stem cells mimicked the effects observed in vivo. Through immunodepletion studies, we identified vascular endothelial growth factor, thrombospondins 1 and 2, and slit as mediators partially responsible for stem cell-induced effects on dendritic sprouting, axonal plasticity and axonal transport in vitro. Thus, we postulate that human neural progenitor cells aid recovery after stroke through secretion of factors that enhance brain repair and plasticity.


Asunto(s)
Transporte Axonal/fisiología , Isquemia Encefálica/cirugía , Corteza Cerebral/citología , Células-Madre Neurales , Plasticidad Neuronal/fisiología , Análisis de Varianza , Animales , Biotina/análogos & derivados , Biotina/metabolismo , Infarto Encefálico/etiología , Infarto Encefálico/patología , Isquemia Encefálica/complicaciones , Supervivencia Celular , Células Cultivadas , Cuerpo Calloso/patología , Dendritas/fisiología , Dextranos/metabolismo , Modelos Animales de Enfermedad , Feto , Regulación de la Expresión Génica , Humanos , Masculino , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Células-Madre Neurales/trasplante , Desempeño Psicomotor/fisiología , ARN Mensajero/metabolismo , Ratas , Ratas Desnudas , Ratas Sprague-Dawley , Factores de Tiempo , Vibrisas/inervación
12.
Exp Neurol ; 210(2): 602-7, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18207142

RESUMEN

A number of gene therapy approaches have been developed for protecting neurons from necrotic neurological insults. Such therapies are limited by the need for transcription and translation of the protective protein, delaying therapeutic impact. As an alternative, we explore the neuroprotective potential of protein therapy, using a fusion protein comprised of the death-suppressing BH4 domain of the Bcl-xL protein and the protein transduction domain of the human immunodeficiency virus Tat protein. This fusion protein decreased neurotoxicity caused by the excitotoxins glutamate and kainic acid in primary hippocampal cultures, and decreased hippocampal damage in vivo in an excitotoxic seizure model.


Asunto(s)
Apoptosis/fisiología , Productos del Gen tat/uso terapéutico , Síndromes de Neurotoxicidad/tratamiento farmacológico , Proteínas Recombinantes/uso terapéutico , Proteína bcl-X/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Interacciones Farmacológicas , Embrión de Mamíferos , Productos del Gen tat/biosíntesis , Ácido Glutámico/toxicidad , Hipocampo/citología , Ácido Kaínico/toxicidad , Masculino , Neuroglía/efectos de los fármacos , Neuronas/efectos de los fármacos , Síndromes de Neurotoxicidad/etiología , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/biosíntesis , Proteína bcl-X/metabolismo
13.
Brain Res ; 1182: 138-43, 2007 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-17950261

RESUMEN

Brain injury due to seizure induces a robust inflammatory response that involves multiple factors. Although the expression of chemokines has been identified as a part of this response, there are remaining questions about their relative contribution to seizure pathogenesis. To address this, we report the expression profile of the chemokine, monocyte chemoattractant protein-1 (MCP-1, CCL2), during kainate-induced seizure in the rat hippocampus. Furthermore, we compare MCP-1 expression to the temporal profile of blood-brain barrier (BBB) permeability and immune cell recruitment at the injury site, since both of these events have been linked to MCP-1. We find that BBB permeability increased prior to upregulation of MCP-1, while MCP-1 upregulation and immune cell recruitment occurred concurrently, 7-13 h after opening of the BBB. Our findings support the following conclusions: (1) BBB opening to large proteins does not require MCP-1 upregulation; (2) Leukocyte immigration is not sufficient to induce BBB opening to large proteins; (3) MCP-1 upregulation likely mediates recruitment of macrophages/microglia and granulocytes during seizure injury, thus warranting further investigation of this chemokine.


Asunto(s)
Quimiocina CCL2/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Hipocampo/efectos de los fármacos , Ácido Kaínico , Estado Epiléptico , Análisis de Varianza , Animales , Antígeno CD11b/metabolismo , Modelos Animales de Enfermedad , Azul de Evans , Hipocampo/metabolismo , Hipocampo/patología , Masculino , Ratas , Ratas Sprague-Dawley , Estado Epiléptico/inducido químicamente , Estado Epiléptico/metabolismo , Estado Epiléptico/patología , Factores de Tiempo
14.
J Biol Chem ; 280(15): 14413-9, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15710616

RESUMEN

DNA methylation within the promoter region of human LINE1 (L1) transposable elements is important for maintaining transcriptional inactivation and for inhibiting L1 transposition. Determining methylation patterns on the complementary strands of repeated sequences is difficult using standard bisulfite methylation analysis. Evolutionary changes in each repeat and the variations between cells or alleles of the same repeat lead to a heterogeneous population of sequences. Potential sequence biases can arise during analyses that are different for the converted sense and antisense strands. These problems can be avoided with hairpin-bisulfite PCR, a double-stranded PCR method in which complementary strands of individual molecules are attached by a hairpin linker ligated to genomic DNA. Using human L1 elements to study methylation of repeated sequences, (i) we distinguish valid L1 sequences from redundant and contaminant sequences by applying the powerful new method of molecular barcodes, (ii) we resolve a controversy on the level of hemimethylation of L1 sequences in fetal fibroblasts in favor of relatively little hemimethylation, (iii) we report that human L1 sequences in different cell types also have primarily concordant CpG methylation patterns on complementary strands, and (iv) we provide evidence that non-CpG cytosines within the regions analyzed are rarely methylated.


Asunto(s)
Islas de CpG , Metilación de ADN , Elementos de Nucleótido Esparcido Largo , Secuencia de Bases , Citosina/química , Evolución Molecular , Femenino , Fibroblastos/metabolismo , Humanos , Datos de Secuencia Molecular , Oligonucleótidos Antisentido/química , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Homología de Secuencia de Ácido Nucleico , Sulfitos/metabolismo , Sulfitos/farmacología
15.
Proc Natl Acad Sci U S A ; 101(1): 204-9, 2004 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-14673087

RESUMEN

Epigenetic inheritance, the transmission of gene expression states from parent to daughter cells, often involves methylation of DNA. In eukaryotes, cytosine methylation is a frequent component of epigenetic mechanisms. Failure to transmit faithfully a methylated or an unmethylated state of cytosine can lead to altered phenotypes in plants and animals. A central unresolved question in epigenetics concerns the mechanisms by which a locus maintains, or changes, its state of cytosine methylation. We developed "hairpin-bisulfite PCR" to analyze these mechanisms. This method reveals the extent of methylation symmetry between the complementary strands of individual DNA molecules. Using hairpin-bisulfite PCR, we determined the fidelity of methylation transmission in the CpG island of the FMR1 gene in human lymphocytes. For the hypermethylated CpG island of this gene, characteristic of inactive-X alleles, we estimate a maintenance methylation efficiency of approximately 0.96 per site per cell division. For de novo methylation efficiency (E(d)), remarkably different estimates were obtained for the hypermethylated CpG island (E(d) = 0.17), compared with the hypomethylated island on the active-X chromosome (E(d) < 0.01). These results clarify the mechanisms by which the alternative hypomethylated and hypermethylated states of CpG islands are stably maintained through many cell divisions. We also analyzed a region of human L1 transposable elements. These L1 data provide accurate methylation patterns for the complementary strand of each repeat sequence analyzed. Hairpin-bisulfite PCR will be a powerful tool in studying other processes for which genetic or epigenetic information differs on the two complementary strands of DNA.


Asunto(s)
Metilación de ADN , ADN/química , ADN/genética , Reacción en Cadena de la Polimerasa/métodos , Proteínas de Unión al ARN , Alelos , Secuencia de Bases , Cromosomas Humanos X/genética , Islas de CpG , Citosina/química , Elementos Transponibles de ADN/genética , Compensación de Dosificación (Genética) , Epigénesis Genética , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , Humanos , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Conformación de Ácido Nucleico , Homología de Secuencia de Ácido Nucleico , Sulfitos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...