Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Foods ; 13(9)2024 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-38731732

RESUMEN

Reducing production costs, known as scaling, is a significant obstacle in the advancement of cultivated meat. The cultivation process hinges on several key components, e.g., cells, media, scaffolds, and bioreactors. This study demonstrates an innovative approach, departing from traditional stainless steel or glass bioreactors, by integrating food-grade plant-based scaffolds and thermoplastic film bioreactors. While thermoplastic films are commonly used for constructing fluidic systems, conventional welding methods are cost-prohibitive and lack rapid prototyping capabilities, thus inflating research and development expenses. The developed laser welding technique facilitates contamination-free and leakproof sealing of polyethylene films, enabling the efficient fabrication of macrofluidic systems with various designs and dimensions. By incorporating food-grade plant-based scaffolds, such as rice seeded with bovine mesenchymal stem cells, into these bioreactors, this study demonstrates sterile cell proliferation on scaffolds within macrofluidic systems. This approach not only reduces bioreactor prototyping and construction costs but also addresses the need for scalable solutions in both research and industrial settings. Integrating single-use bioreactors with minimal shear forces and incorporating macro carriers such as puffed rice may further enhance biomass production in a scaled-out model. The use of food-grade plant-based scaffolds aligns with sustainable practices in tissue engineering and cultured-meat production, emphasizing its suitability for diverse applications.

2.
Mob DNA ; 15(1): 4, 2024 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-38468276

RESUMEN

BACKGROUND: Moloney murine leukemia virus (MLV) replication is suppressed in mouse embryonic stem cells (ESCs) by the Trim28-SETDB1 complex. The chromatin remodeler Smarcad1 interacts with Trim28 and was suggested to allow the deposition of the histone variant H3.3. However, the role of Trim28, H3.3, and Smarcad1 in MLV repression in ESCs still needs to be fully understood. RESULTS: In this study, we used MLV to explore the role of Smarcad1 in retroviral silencing in ESCs. We show that Smarcad1 is immediately recruited to the MLV provirus. Based on the repression dynamics of a GFP-reporter MLV, our findings suggest that Smarcad1 plays a critical role in the establishment and maintenance of MLV repression, as well as other Trim28-targeted genomic loci. Furthermore, Smarcad1 is important for stabilizing and strengthening Trim28 binding to the provirus over time, and its presence around the provirus is needed for proper deposition of H3.3 on the provirus. Surprisingly, the combined depletion of Smarcad1 and Trim28 results in enhanced MLV derepression, suggesting that these two proteins may also function independently to maintain repressive chromatin states. CONCLUSIONS: Overall, the results of this study provide evidence for the crucial role of Smarcad1 in the silencing of retroviral elements in embryonic stem cells. Further research is needed to fully understand how Smarcad1 and Trim28 cooperate and their implications for gene expression and genomic stability.

3.
iScience ; 26(8): 107305, 2023 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-37529103

RESUMEN

The adverse effects of heat stress (HS) on physiological systems are well documented, yet the underlying molecular mechanisms behind it remain poorly understood. To address this knowledge gap, we conducted a comprehensive investigation into the impact of HS on mesenchymal stem cells (MSCs), focusing on their morphology, phenotype, proliferative capacity, and fate determination. Our in-depth analysis of the MSCs' transcriptome revealed a significant influence of HS on the transcriptional landscape. Notably, even after a short period of stress, we observed a persistent alteration in cell identity, potentially mediated by the activation of bivalent genes. Furthermore, by comparing the differentially expressed genes following short HS with their transcriptional state after recovery, we identified the transient upregulation of MLL and other histone modifiers, providing a potential mechanistic explanation for the stable activation of bivalent genes. This could be used to predict and modify the long-term effect of HS on cell identity.

4.
Clin Epigenetics ; 15(1): 83, 2023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-37170146

RESUMEN

BACKGROUND: Integration of retroviruses into the host genome can impair the genomic and epigenomic integrity of the cell. As a defense mechanism, epigenetic modifications on the proviral DNA repress retroviral sequences in mouse embryonic stem cells (ESC). Here, we focus on the histone 3 variant H3.3, which is abundant in active transcription zones, as well as centromeres and heterochromatinized repeat elements, e.g., endogenous retroviruses (ERV). RESULTS: To understand the involvement of H3.3 in the epigenetic silencing of retroviral sequences in ESC, we depleted the H3.3 genes in ESC and transduced the cells with GFP-labeled MLV pseudovirus. This led to altered retroviral repression and reduced Trim28 recruitment, which consequently led to a loss of heterochromatinization in proviral sequences. Interestingly, we show that H3.3 depletion has a differential effect depending on which of the two genes coding for H3.3, H3f3a or H3f3b, are knocked out. Depletion of H3f3a resulted in a transient upregulation of incoming retroviral expression and ERVs, while the depletion of H3f3b did not have the same effect and repression was maintained. However, the depletion of both genes resulted in a stable activation of the retroviral promoter. These findings suggest that H3.3 is important for regulating retroviral gene expression in mouse ESC and provide evidence for a distinct function of the two H3.3 genes in this regulation. Furthermore, we show that Trim28 is needed for depositing H3.3 in retroviral sequences, suggesting a functional interaction between Trim28 recruitment and H3.3 loading. CONCLUSIONS: Identifying the molecular mechanisms by which H3.3 and Trim28 interact and regulate retroviral gene expression could provide a deeper understanding of the fundamental processes involved in retroviral silencing and the general regulation of gene expression, thus providing new answers to a central question of stem cell biology.


Asunto(s)
Retrovirus Endógenos , Histonas , Animales , Ratones , Histonas/genética , Histonas/metabolismo , Proteínas Nucleares/genética , Proteínas Represoras/genética , Metilación de ADN , Silenciador del Gen , Células Madre Embrionarias/metabolismo , Retrovirus Endógenos/genética , Retrovirus Endógenos/metabolismo , Provirus/genética
5.
BMC Vet Res ; 19(1): 64, 2023 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-36997964

RESUMEN

The use of mesenchymal stromal cells (MSCs) is emerging as an efficacious and safe treatment for many infectious and non-infectious inflammatory diseases in human and veterinary medicine. Such use could be done to treat mastitis and metritis, which are the most common disease conditions affecting dairy cows leading to considerable economic losses and reduced animal welfare. Currently, both disease conditions are commonly treated using local and systemic administration of antibiotics. However, this strategy has many disadvantages including low cure rates and the public health hazards. Looking for alternative approaches, we investigated the properties of MSCs using in-vitro mammary and endometrial cell systems and in-vivo mastitis and metritis murine model systems. In-vitro, co-culture of mammary and uterus epithelial cells constructed with NF-kB reporter system, the master regulator of inflammation, demonstrated their anti-inflammatory effects in response to.LPS. In vivo, we challenge animals with field strains of mammary and utero pathogenic Escherichia coli and evaluated the effects of local and systemic application of MSC in the animal models. Disease outcome was evaluated using histological analysis, bacterial counts and gene expression of inflammatory markers. We show that MSC treatment reduced bacterial load in metritis and significantly modulated the inflammatory response of the uterus and mammary gland to bacterial infection. Most notably are the immune modulatory effects of remotely engrafted intravenous MSCs, which open new avenues to the development of MSC-based cell-free therapies.


Asunto(s)
Enfermedades de los Bovinos , Mastitis Bovina , Células Madre Mesenquimatosas , Femenino , Bovinos , Humanos , Animales , Ratones , Escherichia coli , Inflamación/veterinaria , Inflamación/patología , Útero/patología , Glándulas Mamarias Animales/patología , Mastitis Bovina/microbiología , Enfermedades de los Bovinos/patología
6.
EMBO Rep ; 23(9): e55101, 2022 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-35972201

RESUMEN

Emerging evidence shows that transposable elements (TEs) are induced in response to viral infections. This TE induction is suggested to trigger a robust and durable interferon response, providing a host defense mechanism. Here, we analyze TE expression changes in response to SARS-CoV-2 infection in different human cellular models. Unlike other viruses, SARS-CoV-2 infection does not lead to global upregulation of TEs in primary cells. We report a correlation between TEs activation and induction of interferon-related genes, suggesting that failure to activate TEs may account for the weak interferon response. Moreover, we identify two variables that explain most of the observed diverseness in immune responses: basal expression levels of TEs in the pre-infected cells and the viral load. Finally, analyzing the SARS-CoV-2 interactome and the epigenetic landscape around the TEs activated following infection, we identify SARS-CoV-2 interacting proteins, which may regulate chromatin structure and TE transcription. This work provides a possible functional explanation for SARS-CoV-2 success in its fight against the host immune system and suggests that TEs could serve as potential drug targets for COVID-19.


Asunto(s)
COVID-19 , Antivirales , COVID-19/genética , Elementos Transponibles de ADN/genética , Humanos , Interferones/genética , SARS-CoV-2
7.
Int J Mol Sci ; 23(10)2022 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-35628565

RESUMEN

Mesenchymal stem cells (MSC) have many roles that are important for the body's proper functioning. When the MSC pool is damaged, it is often correlated with impaired development or health of the organism. MSC are known for their anti-inflammatory, immunomodulatory and trophic characteristics that play an important role in the physiological homeostasis of many tissues. Heat shock impairs MSC capacity by inducing the generation of reactive oxygen species and mitochondrial dysfunction, which, in turn, send the cells into a state of premature senescence. Here, we pre-exposed MSC to melatonin, resveratrol, or curcumin, which are natural antioxidative compounds, and tested the protective effects of these substances from oxidative stress and aging. Our data showed that pre-exposure of MSC to antioxidants decreased reactive oxygen species while mitochondrial damage remained high. Additionally, although the proliferation of the cells was slow, antioxidants protected the cells from premature senescence, and subsequent cytokine release was prevented. We conclude that while elevated temperatures directly cause mitochondrial damage, senescence is induced by elevated ROS levels. We suggest that heat shock alters cell and tissue homeostasis by several independent mechanisms; however, reducing tissue senescence will reduce damage and provide a pathway to overcome physiological challenges in animals.


Asunto(s)
Antioxidantes , Células Madre Mesenquimatosas , Animales , Antioxidantes/metabolismo , Antioxidantes/farmacología , Bovinos , Senescencia Celular , Respuesta al Choque Térmico , Células Madre Mesenquimatosas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
8.
Front Cell Dev Biol ; 8: 565970, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33072750

RESUMEN

Heat stress can have a serious impact on the health of both humans and animals. A major question is how heat stress affects normal development and differentiation at both the cellular and the organism levels. Here we use an in vitro experimental system to address how heat shock treatment influences the properties of bovine mesenchymal stem cells (MSCs)-multipotent progenitor cells-which are found in most tissues. Because cattle are sensitive to harsh external temperatures, studying the effects of heat shock on MSCs provides a unique platform to address cellular stress in a physiologically relevant model organism. Following isolation and characterization of MSCs from the cow's umbilical cord, heat shock was induced either as a pulse (1 h) or continuously (3 days), and consequent effects on MSCs were characterized. Heat shock induced extensive phenotypic changes in MSCs and dramatically curtailed their capacity to proliferate and differentiate. These changes were associated with a partial arrest in the G1/S or G2/M checkpoints. Furthermore, MSCs lost their ability to resolve the inflammatory response of RAW macrophages in coculture. A possible explanation for this loss of function is the accumulation of reactive oxygen species and malfunction of the mitochondria in the treated cells. Heat shock treatments resulted in stress-induced premature senescence, affecting the MSCs' ability to proliferate properly for many cell passages to follow. Exposure to elevated external temperatures leads to mitochondrial damage and oxidative stress, which in turn conveys critical changes in the proliferation, differentiation, and immunomodulatory phenotype of heat-stressed MSCs. A better understanding of the effect of heat shock on humans and animals may result in important health and economic benefits.

9.
Viruses ; 12(9)2020 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-32932986

RESUMEN

Embryonic stem cells (ESC) have the ability to epigenetically silence endogenous and exogenous retroviral sequences. Trim28 plays an important role in establishing this silencing, but less is known about the role other Trim proteins play. The Tif1 family is a sub-group of the Trim family, which possess histone binding ability in addition to the distinctive RING domain. Here, we have examined the interaction between three Tif1 family members, namely Trim24, Trim28 and Trim33, and their function in retroviral silencing. We identify a complex formed in ESC, comprised of these three proteins. We further show that when Trim33 is depleted, the complex collapses and silencing efficiency of both endogenous and exogenous sequences is reduced. Similar transcriptional activation takes place when Trim24 is depleted. Analysis of the H3K9me3 chromatin modification showed a decrease in this repressive mark, following both Trim24 and Trim33 depletion. As Trim28 is an identified binding partner of the H3K9 methyltransferase ESET, this further supports the involvement of Trim28 in the complex. The results presented here suggest that a complex of Tif1 family members, each of which possesses different specificity and efficiency, contributes to the silencing of retroviral sequences in ESC.


Asunto(s)
Proteínas Portadoras/metabolismo , Células Madre Embrionarias/metabolismo , Epigénesis Genética/fisiología , Silenciador del Gen , Retroviridae/genética , Retroviridae/fisiología , Factores de Transcripción/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Portadoras/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/virología , Retrovirus Endógenos , Células HEK293 , Histonas/metabolismo , Humanos , Virus de la Leucemia Murina , Ratones , Células 3T3 NIH , Proteínas Nucleares , Unión Proteica , Factores de Transcripción/genética , Proteína 28 que Contiene Motivos Tripartito/metabolismo
10.
Dev Cell ; 48(2): 135-150, 2019 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-30695696

RESUMEN

Pluripotent embryonic stem cells (ESCs) are considered to have open and accessible chromatin relative to differentiated cells. However, as many studies supporting these conclusions relied on ESCs grown in serum, it has been suggested that some of these features are the result of culture conditions, particularly as more recent work using GSK3/MEK inhibitors ("2i") to mimic "ground-state" conditions of the pre-implantation blastocyst observed some altered epigenetic features. Here, we systematically review chromatin and epigenetic features in 2i- and serum-grown conditions to come to a clearer picture of what are genuine characteristics of pluripotency and what open chromatin features predict pluripotency.


Asunto(s)
Diferenciación Celular/fisiología , Cromatina/metabolismo , Células Madre Embrionarias/citología , Células Madre Pluripotentes/citología , Animales , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Ratones , Células Madre Embrionarias de Ratones/citología
11.
Nucleic Acids Res ; 45(21): 12181-12194, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-29036702

RESUMEN

Histone variants and their chaperones are key regulators of eukaryotic transcription, and are critical for normal development. The histone variant H3.3 has been shown to play important roles in pluripotency and differentiation, and although its genome-wide patterns have been investigated, little is known about the role of its dynamic turnover in transcriptional regulation. To elucidate the role of H3.3 dynamics in embryonic stem cell (ESC) biology, we generated mouse ESC lines carrying a single copy of a doxycycline (Dox)-inducible HA-tagged version of H3.3 and monitored the rate of H3.3 incorporation by ChIP-seq at varying time points following Dox induction, before and after RA-induced differentiation. Comparing H3.3 turnover profiles in ESCs and RA-treated cells, we identified a hyperdynamic H3.3-containing nucleosome at the -1 position in promoters of genes expressed in ESCs. This dynamic nucleosome is restricted and shifted downstream into the +1 position following differentiation. We suggest that histone turnover dynamics provides an additional mechanism involved in expression regulation, and that a hyperdynamic -1 nucleosome marks promoters in ESCs. Our data provide evidence for regional regulation of H3.3 turnover in ESC promoters, and calls for testing, in high resolution, the dynamic behavior of additional histone variants and other structural chromatin proteins.


Asunto(s)
Células Madre Embrionarias/metabolismo , Código de Histonas , Histonas/metabolismo , Nucleosomas/metabolismo , Regiones Promotoras Genéticas , Animales , Células Cultivadas , Inmunoprecipitación de Cromatina , Elementos de Facilitación Genéticos , Ratones , Sitio de Iniciación de la Transcripción , Transcripción Genética
12.
Cell ; 163(1): 230-45, 2015 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-26365490

RESUMEN

Embryonic stem cells (ESCs) repress the expression of exogenous proviruses and endogenous retroviruses (ERVs). Here, we systematically dissected the cellular factors involved in provirus repression in embryonic carcinomas (ECs) and ESCs by a genome-wide siRNA screen. Histone chaperones (Chaf1a/b), sumoylation factors (Sumo2/Ube2i/Sae1/Uba2/Senp6), and chromatin modifiers (Trim28/Eset/Atf7ip) are key determinants that establish provirus silencing. RNA-seq analysis uncovered the roles of Chaf1a/b and sumoylation modifiers in the repression of ERVs. ChIP-seq analysis demonstrates direct recruitment of Chaf1a and Sumo2 to ERVs. Chaf1a reinforces transcriptional repression via its interaction with members of the NuRD complex (Kdm1a, Hdac1/2) and Eset, while Sumo2 orchestrates the provirus repressive function of the canonical Zfp809/Trim28/Eset machinery by sumoylation of Trim28. Our study reports a genome-wide atlas of functional nodes that mediate proviral silencing in ESCs and illuminates the comprehensive, interconnected, and multi-layered genetic and epigenetic mechanisms by which ESCs repress retroviruses within the genome.


Asunto(s)
Células Madre Embrionarias/virología , Retrovirus Endógenos/genética , Provirus/genética , Animales , Factor 1 de Ensamblaje de la Cromatina/genética , Factor 1 de Ensamblaje de la Cromatina/metabolismo , Células Madre de Carcinoma Embrionario/virología , Epigénesis Genética , Ratones , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo
13.
Mol Cell Biol ; 35(5): 770-7, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25547290

RESUMEN

Retroviruses have evolved complex transcriptional enhancers and promoters that allow their replication in a wide range of tissue and cell types. Embryonic stem (ES) cells, however, characteristically suppress transcription of proviruses formed after infection by exogenous retroviruses and also of most members of the vast array of endogenous retroviruses in the genome. These cells have unusual profiles of transcribed genes and are poised to make rapid changes in those profiles upon induction of differentiation. Many of the transcription factors in ES cells control both host and retroviral genes coordinately, such that retroviral expression patterns can serve as markers of ES cell pluripotency. This overlap is not coincidental; retrovirus-derived regulatory sequences are often used to control cellular genes important for pluripotency. These sequences specify the temporal control and perhaps "noisy" control of cellular genes that direct proper cell gene expression in primitive cells and their differentiating progeny. The evidence suggests that the viral elements have been domesticated for host needs, reflecting the wide-ranging exploitation of any and all available DNA sequences in assembling regulatory networks.


Asunto(s)
Células Madre Embrionarias/virología , Retrovirus Endógenos/genética , Silenciador del Gen , Células Madre Pluripotentes/virología , Animales , Cromatina/química , Elementos Transponibles de ADN , Células Madre Embrionarias/citología , Epigénesis Genética , Regulación Viral de la Expresión Génica , Redes Reguladoras de Genes , Genoma Viral , Heterocromatina/química , Humanos , Ratones , Células Madre Pluripotentes/citología , Análisis de Secuencia de ADN , Factores de Transcripción/metabolismo
14.
Retrovirology ; 11: 31, 2014 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-24742368

RESUMEN

BACKGROUND: Retroviral DNAs are profoundly silenced at the transcriptional level in embryonic cell types. The transcriptional profile of pluripotent stem cells has been demonstrated to be extremely heterogeneous from cell to cell, and how the silencing of retroviral DNAs is achieved is not yet well characterized. RESULTS: In the current study, we investigated the transcriptional silencing dynamics in stem cells by independently monitoring the expression of two Moloney murine leukemia virus (MMLV) retroviral vectors newly introduced into embryonic carcinoma (EC) cells. Although MMLV is efficiently silenced by epigenetic mechanisms in most such cells, a small number of the doubly-transduced EC cells transiently show double-positive proviral expression. These cells were sorted and their expression patterns were studied over time as silencing is established. CONCLUSIONS: Our data suggest that retroviral silencing occurs stochastically, in an individual locus-specific fashion, and often without synchronous silencing of both viruses in the same cells. Surprisingly, the chromatin modifications that mark the silenced proviruses are unchanged even in cells that temporarily escape silencing. This local silencing effect is a feature of stem cell epigenomic regulation that has not previously been revealed.


Asunto(s)
Células Madre Embrionarias/fisiología , Silenciador del Gen , Genoma Viral/genética , Retroviridae/genética , Animales , Línea Celular , ADN Viral/genética , Epigénesis Genética/genética , Vectores Genéticos , Ratones , Virus de la Leucemia Murina de Moloney/genética , Células 3T3 NIH , Transcripción Genética
15.
Cell Rep ; 4(1): 50-8, 2013 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-23810560

RESUMEN

Embryonic cells transcriptionally repress the expression of endogenous and exogenous retroelements. Trim28, a key player in this silencing, is known to act in a large DNA-bound complex, but the other components of the complex are not fully characterized. Here, we show that the zinc finger protein Yin Yang 1 (YY1) is one such component. YY1 binds to the long terminal repeat (LTR) region of both exogenous and endogenous retroviruses (ERVs). Deletion of the YY1-binding site from the retroviral genome leads to a major loss of silencing in embryonic cells and a coordinated loss of repressive histone marks from the proviral chromatin. Depletion of YY1 protein results in marked upregulation of expression of exogenous viruses and of selected ERVs. Finally, we report an embryonic cell-specific interaction between YY1 and Trim28. Our results suggest a major role for YY1 in the silencing of both exogenous retroviruses and ERVs in embryonic cells.


Asunto(s)
Células Madre Embrionarias/metabolismo , Silenciador del Gen , Provirus/genética , Factor de Transcripción YY1/metabolismo , Animales , Sitios de Unión , Cromatina/metabolismo , ADN Viral/genética , ADN Viral/metabolismo , Células Madre Embrionarias/virología , Eliminación de Gen , Células HEK293 , Histonas/metabolismo , Humanos , Ratones , Virus de la Leucemia Murina de Moloney/genética , Virus de la Leucemia Murina de Moloney/metabolismo , Células 3T3 NIH , Proteínas Nucleares/metabolismo , Provirus/metabolismo , Proteínas Represoras/metabolismo , Secuencias Repetidas Terminales , Transcripción Genética , Proteína 28 que Contiene Motivos Tripartito , Factor de Transcripción YY1/genética
16.
EMBO Rep ; 14(1): 73-9, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23154467

RESUMEN

Embryonic stem cells repress retroviral infection through transcriptional silencing of proviral DNAs. We characterized two distinct mechanisms of silencing in embryonic mouse cells infected by Moloney murine leukaemia virus (MLV): a highly efficient one targeting the proline transfer RNA primer-binding site (PBSpro), and a less efficient one operating independently of the PBS. Rare virus-expressing populations were isolated, and the timing and efficiency of establishment of silencing were determined. Superinfection of the selected virus-expressing cells with a second virus carrying a distinguishable reporter revealed that the PBSpro-directed silencing was still largely intact, whereas the PBS-independent silencing was partially reduced. The timing and stability of silencing, and the associated chromatin modifications on newly established and endogenous proviruses were determined. The results indicate that epigenetic mechanisms with different specificity and efficiency are used to silence the exogenous retroviral sequences in embryonic cells.


Asunto(s)
ADN Viral/genética , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/virología , Epigénesis Genética , Virus de la Leucemia Murina de Moloney/genética , Provirus/genética , Animales , Cromatina/genética , Cromatina/metabolismo , Metilación de ADN , ADN Viral/antagonistas & inhibidores , Células Madre Embrionarias/citología , Silenciador del Gen , Genes Reporteros , Proteínas Fluorescentes Verdes , Interacciones Huésped-Patógeno , Humanos , Ratones , Células 3T3 NIH , ARN Interferente Pequeño/genética , ARN de Transferencia de Prolina/genética , ARN de Transferencia de Prolina/metabolismo , Transducción de Señal
17.
Nature ; 490(7421): 561-5, 2012 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-23023124

RESUMEN

Although most genes are expressed biallelically, a number of key genomic sites--including immune and olfactory receptor regions--are controlled monoallelically in a stochastic manner, with some cells expressing the maternal allele and others the paternal allele in the target tissue. Very little is known about how this phenomenon is regulated and programmed during development. Here, using mouse immunoglobulin-κ (Igκ) as a model system, we demonstrate that although individual haematopoietic stem cells are characterized by allelic plasticity, early lymphoid lineage cells become committed to the choice of a single allele, and this decision is then stably maintained in a clonal manner that predetermines monoallelic rearrangement in B cells. This is accompanied at the molecular level by underlying allelic changes in asynchronous replication timing patterns at the κ locus. These experiments may serve to define a new concept of stem cell plasticity.


Asunto(s)
Alelos , Linaje de la Célula , Reordenamiento Génico de Cadena Ligera de Linfocito B/genética , Cadenas kappa de Inmunoglobulina/genética , Células Precursoras de Linfocitos B/citología , Células Precursoras de Linfocitos B/metabolismo , Animales , Inmunoprecipitación de Cromatina , Células Clonales/citología , Células Clonales/inmunología , Células Clonales/metabolismo , Momento de Replicación del ADN , Femenino , Hematopoyesis , Humanos , Cadenas kappa de Inmunoglobulina/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Modelos Animales , Modelos Inmunológicos , Células Precursoras de Linfocitos B/inmunología , Procesos Estocásticos
18.
Genes Dev ; 23(20): 2437-47, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19833769

RESUMEN

Human cells contain several hundred ribosomal genes (rDNA) that are clustered into nucleolar organizer regions (NORs) on the short arms of five different acrocentric chromosomes. Only approximately 50% of the gene copies are actually expressed in somatic cells. Here, we used a new cytological technique to demonstrate that rDNA is regulated allelically in a regional manner, with one parental copy of each NOR being repressed in any individual cell. This process is similar to that of X-chromosome inactivation in females. Early in development, one copy of each NOR becomes late-replicating, thus probably marking it for inactivation and subsequent targeted de novo methylation at rDNA promoter regions. Once established, this multichromosomal allelic pattern is then maintained clonally in somatic cells. This pathway may serve as an epigenetic mechanism for controlling the number of available rDNA copies during development.


Asunto(s)
Alelos , ADN Ribosómico/genética , Silenciador del Gen , Animales , Línea Celular , Metilación de ADN , Momento de Replicación del ADN , Regulación del Desarrollo de la Expresión Génica , Genes de ARNr/genética , Histonas/metabolismo , Humanos , Ratones , Hibridación de Ácido Nucleico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...