Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Mol Biosci ; 10: 1286690, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38143802

RESUMEN

Metabolic chemical reporters (MCRs) provide easily accessible means to study glycans in their native environments. However, because monosaccharide precursors are shared by many glycosylation pathways, selective incorporation has been difficult to attain. Here, a strategy for defining the selectivity and enzymatic incorporation of an MCR is presented. Performing ß-elimination to interrogate O-linked sugars and using commercially available glycosidases and glycosyltransferase inhibitors, we probed the specificity of widely used azide (Ac4GalNAz) and alkyne (Ac4GalNAlk and Ac4GlcNAlk) sugar derivatives. Following the outlined strategy, we provide a semiquantitative assessment of the specific and non-specific incorporation of this bioorthogonal sugar (Ac4GalNAz) into numerous N- and O-linked glycosylation pathways. This approach should be generally applicable to other MCRs to define the extent of incorporation into the various glycan species.

2.
Res Sq ; 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37577573

RESUMEN

Metabolic incorporation of chemically tagged monosaccharides is a facile means of labelling cellular glycoprotein and glycolipids. Yet, since the monosaccharide precursors are often shared by several pathways, selectivity has been difficult to attain. For example, N-linked glycosylation is a chemically complex, and ubiquitous post translational modification with three distinct classes of GlcNAc-containing N-glycan structures: oligomannose, hybrid, and complex. Here we describe synthesis of 1,3-Pr2-6-OTs GlcNAlk as a next generation metabolic chemical reporter (MCR) for the specific labeling of hybrid N-glycan structures. We first developed a general strategy for defining the selectivity of labelling with chemically tagged monosaccharides. We then applied this approach to establish that 1,3-Pr2-6-OTs GlcNAlk is specifically incorporated into hybrid N-glycans. Using this MCR as a detection tool, we carried out imaging experiments to define the intracellular localization and trafficking of target proteins bearing hybrid N-glycan structures.

3.
PLoS One ; 17(9): e0274076, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36112613

RESUMEN

Genetic and environmental manipulations, such as dietary restriction, can improve both health span and lifespan in a wide range of organisms, including humans. Changes in nutrient intake trigger often overlapping metabolic pathways that can generate distinct or even opposite outputs depending on several factors, such as when dietary restriction occurs in the lifecycle of the organism or the nature of the changes in nutrients. Due to the complexity of metabolic pathways and the diversity in outputs, the underlying mechanisms regulating diet-associated pro-longevity are not yet well understood. Adult reproductive diapause (ARD) in the model organism Caenorhabditis elegans is a dietary restriction model that is associated with lengthened lifespan and reproductive potential. To explore the metabolic pathways regulating ARD in greater depth, we performed a candidate-based genetic screen analyzing select nutrient-sensing pathways to determine their contribution to the regulation of ARD. Focusing on the three phases of ARD (initiation, maintenance, and recovery), we found that ARD initiation is regulated by fatty acid metabolism, sirtuins, AMPK, and the O-linked N-acetyl glucosamine (O-GlcNAc) pathway. Although ARD maintenance was not significantly influenced by the nutrient sensors in our screen, we found that ARD recovery was modulated by energy sensing, stress response, insulin-like signaling, and the TOR pathway. Further investigation of downstream targets of NHR-49 suggest the transcription factor influences ARD initiation through the fatty acid ß-oxidation pathway. Consistent with these findings, our analysis revealed a change in levels of neutral lipids associated with ARD entry defects. Our findings identify conserved genetic pathways required for ARD entry and recovery and uncover genetic interactions that provide insight into the role of OGT and OGA.


Asunto(s)
Diapausa , Nutrientes , Transducción de Señal , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Caenorhabditis elegans/metabolismo , Diapausa/genética , Diapausa/fisiología , Ácidos Grasos/metabolismo , Glucosamina/metabolismo , Humanos , Insulinas/metabolismo , Lípidos/química , Nutrientes/metabolismo , Nutrientes/farmacología , Reproducción/genética , Reproducción/fisiología , Transducción de Señal/genética , Sirtuinas/genética , Sirtuinas/metabolismo , Factores de Transcripción/metabolismo
4.
Front Immunol ; 13: 802336, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35432339

RESUMEN

Inflammation is the immune response to harmful stimuli, including pathogens, damaged cells and toxic compounds. However, uncontrolled inflammation can be detrimental and contribute to numerous chronic inflammatory diseases, such as insulin resistance. At the forefront of this response are macrophages, which sense the local microenvironment to respond with a pro-inflammatory, M1-polarized phenotype, or anti-inflammatory, M2-polarized phenotype. M1 macrophages upregulate factors like pro-inflammatory cytokines, to promote inflammatory signaling, and inducible Nitric Oxide Synthase (iNOS), to produce nitric oxide (NO). The generated NO can kill microorganisms to protect the body, but also signal back to the macrophage to limit pro-inflammatory cytokine production to maintain macrophage homeostasis. Thus, the tight regulation of iNOS in macrophages is critical for the immune system. Here, we investigated how elevation of the nutrient-sensitive posttranslational modification, O-GlcNAc, impacts M1 polarized macrophages. We identified increased gene expression of specific pro-inflammatory cytokines (Il-6, Il-1ß, Il-12) when O-GlcNAc cycling was blocked. We further uncovered an interaction between O-GlcNAc and iNOS, with iNOS being an OGT target in vitro. Analysis of M1 polarized bone marrow derived macrophages deficient in the enzyme that removes O-GlcNAc, O-GlcNAcase (OGA), revealed decreased iNOS activity as measured by a reduction in NO release. Further, elevated O-GlcNAc acted on Il-6 expression through the iNOS pathway, as iNOS inhibitior L-NIL raised wildtype Il-6 expression similar to OGA deficient cells but had no further effect on the hyper-O-GlcNAcylated cells. Thus O-GlcNAc contributes to macrophage homeostasis through modulation of iNOS activity.


Asunto(s)
Citocinas , Óxido Nítrico , Acetilglucosaminidasa , Citocinas/metabolismo , Humanos , Inflamación/metabolismo , Interleucina-6/metabolismo , Macrófagos/metabolismo , Óxido Nítrico/metabolismo
5.
PLoS Genet ; 18(3): e1010128, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35294432

RESUMEN

Tissue homeostasis requires a delicate balance between stem cell self-renewal, proliferation, and differentiation. Essential to this process is glycosylation, with both intra-and extra-cellular glycosylation being required for stem cell homeostasis. However, it remains unknown how intracellular glycosylation, O-GlcNAcylation, interfaces with cellular components of the extracellular glycosylation machinery, like the cytosolic N-glycanase NGLY1. In this study, we utilize the Drosophila gut and uncover a pathway in which O-GlcNAcylation cooperates with the NGLY1 homologue PNG1 to regulate proliferation in intestinal stem cells (ISCs) and apoptosis in differentiated enterocytes. Further, the CncC antioxidant signaling pathway and ENGase, an enzyme involved in the processing of free oligosaccharides in the cytosol, interact with O-GlcNAc and PNG1 through regulation of protein aggregates to contribute to gut maintenance. These findings reveal a complex coordinated regulation between O-GlcNAcylation and the cytosolic glycanase PNG1 critical to balancing proliferation and apoptosis to maintain gut homeostasis.


Asunto(s)
Apoptosis , Drosophila , Animales , Proliferación Celular , Citosol , Drosophila/metabolismo , Homeostasis
6.
J Biol Chem ; 298(3): 101743, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35183508

RESUMEN

Endomembrane glycosylation and cytoplasmic O-GlcNAcylation each play essential roles in nutrient sensing, and characteristic changes in glycan patterns have been described in disease states such as diabetes and cancer. These changes in glycosylation have important functional roles and can drive disease progression. However, little is known about the molecular mechanisms underlying how these signals are integrated and transduced into biological effects. Galectins are proteins that bind glycans and that are secreted by a poorly characterized nonclassical secretory mechanism. Once outside the cell, galectins bind to the terminal galactose residues of cell surface glycans and modulate numerous extracellular functions, such as clathrin-independent endocytosis (CIE). Originating in the cytoplasm, galectins are predicted substrates for O-GlcNAc addition and removal; and as we have shown, galectin 3 is a substrate for O-GlcNAc transferase. In this study, we also show that galectin 3 secretion is sensitive to changes in O-GlcNAc levels. We determined using immunoprecipitation and Western blotting that there is a significant difference in O-GlcNAcylation status between cytoplasmic and secreted galectin 3. We observed dramatic alterations in galectin 3 secretion in response to nutrient conditions, which were dependent on dynamic O-GlcNAcylation. Importantly, we showed that these O-GlcNAc-driven alterations in galectin 3 secretion also facilitated changes in CIE. These results indicate that dynamic O-GlcNAcylation of galectin 3 plays a role in modulating its secretion and can tune its function in transducing nutrient-sensing information coded in cell surface glycosylation into biological effects.


Asunto(s)
Galectina 3 , Acetilglucosamina/metabolismo , Clatrina/metabolismo , Galectina 3/genética , Galectina 3/metabolismo , Glicosilación , N-Acetilglucosaminiltransferasas/metabolismo , Nutrientes , Polisacáridos/metabolismo , Procesamiento Proteico-Postraduccional
7.
Sci Rep ; 11(1): 22106, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34764359

RESUMEN

O-GlcNAcylation is a prevalent form of glycosylation that regulates proteins within the cytosol, nucleus, and mitochondria. The O-GlcNAc modification can affect protein cellular localization, function, and signaling interactions. The specific impact of O-GlcNAcylation on mitochondrial morphology and function has been elusive. In this manuscript, the role of O-GlcNAcylation on mitochondrial fission, oxidative phosphorylation (Oxphos), and the activity of electron transport chain (ETC) complexes were evaluated. In a cellular environment with hyper O-GlcNAcylation due to the deletion of O-GlcNAcase (OGA), mitochondria showed a dramatic reduction in size and a corresponding increase in number and total mitochondrial mass. Because of the increased mitochondrial content, OGA knockout cells exhibited comparable coupled mitochondrial Oxphos and ATP levels when compared to WT cells. However, we observed reduced protein levels for complex I and II when comparing normalized mitochondrial content and reduced linked activity for complexes I and III when examining individual ETC complex activities. In assessing mitochondrial fission, we observed increased amounts of O-GlcNAcylated dynamin-related protein 1 (Drp1) in cells genetically null for OGA and in glioblastoma cells. Individual regions of Drp1 were evaluated for O-GlcNAc modifications, and we found that this post-translational modification (PTM) was not limited to the previously characterized residues in the variable domain (VD). Additional modification sites are predicted in the GTPase domain, which may influence enzyme activity. Collectively, these results highlight the impact of O-GlcNAcylation on mitochondrial dynamics and ETC function and mimic the changes that may occur during glucose toxicity from hyperglycemia.


Asunto(s)
Acilación/genética , Acilación/fisiología , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/fisiología , N-Acetilglucosaminiltransferasas/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Dinaminas/genética , Dinaminas/metabolismo , Glucosa/genética , Glucosa/metabolismo , Glicosilación , Células HCT116 , Humanos , Ratones , Ratones Noqueados , Mitocondrias Cardíacas/genética , Dinámicas Mitocondriales/genética , Dinámicas Mitocondriales/fisiología , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , N-Acetilglucosaminiltransferasas/genética , Fosforilación Oxidativa , Procesamiento Proteico-Postraduccional/genética , Transducción de Señal/genética
8.
Sci Adv ; 7(17)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33883138

RESUMEN

Nuclear receptors farnesoid X receptor (FXR) and small heterodimer partner (SHP) are key regulators of metabolism. Here, we report a previously unknown function for the hepatic FXR-SHP axis in controlling protein N-linked glycosylation. Transcriptome analysis in liver-specific Fxr-Shp double knockout (LDKO) livers revealed induction of genes encoding enzymes in the N-glycosylation pathway, including Mgat5, Fut8, St3gal6, and St6gal1 FXR activation suppressed Mgat5, while Shp deletion induced St3gal6 and St6gal1 Increased percentages of core-fucosylated and triantennary glycan moieties were seen in LDKO livers, and proteins with the "hyperglycoforms" preferentially localized to exosomes and lysosomes. This up-regulation of N-glycosylation machinery was specific to the Golgi apparatus and not the endoplasmic reticulum. The increased glycan complexity in the LDKO correlated well with dilated unstacked Golgi ribbons and alterations in the secretion of albumin, cholesterol, and triglycerides. Our findings demonstrate a role for the FXR-SHP axis in maintaining glycoprotein diversity in the liver.


Asunto(s)
Hígado , Receptores Citoplasmáticos y Nucleares , Colesterol/metabolismo , Hígado/metabolismo , Polisacáridos/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Receptores Citoplasmáticos y Nucleares/metabolismo , Activación Transcripcional
9.
Front Genet ; 11: 605263, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33329753

RESUMEN

Cellular identity in multicellular organisms is maintained by characteristic transcriptional networks, nutrient consumption, energy production and metabolite utilization. Integrating these cell-specific programs are epigenetic modifiers, whose activity is often dependent on nutrients and their metabolites to function as substrates and co-factors. Emerging data has highlighted the role of the nutrient-sensing enzyme O-GlcNAc transferase (OGT) as an epigenetic modifier essential in coordinating cellular transcriptional programs and metabolic homeostasis. OGT utilizes the end-product of the hexosamine biosynthetic pathway to modify proteins with O-linked ß-D-N-acetylglucosamine (O-GlcNAc). The levels of the modification are held in check by the O-GlcNAcase (OGA). Studies from model organisms and human disease underscore the conserved function these two enzymes of O-GlcNAc cycling play in transcriptional regulation, cellular plasticity and mitochondrial reprogramming. Here, we review these findings and present an integrated view of how O-GlcNAc cycling may contribute to cellular memory and transgenerational inheritance of responses to parental stress. We focus on a rare human genetic disorder where mutant forms of OGT are inherited or acquired de novo. Ongoing analysis of this disorder, OGT- X-linked intellectual disability (OGT-XLID), provides a window into how epigenetic factors linked to O-GlcNAc cycling may influence neurodevelopment.

10.
Cell Rep ; 31(6): 107632, 2020 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-32402277

RESUMEN

Stem/progenitor cells exhibit high proliferation rates, elevated nutrient uptake, altered metabolic flux, and stress-induced genome instability. O-GlcNAcylation is an essential post-translational modification mediated by O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), which act in a nutrient- and stress-responsive manner. The precise role of O-GlcNAc in adult stem cells and the relationship between O-GlcNAc and the DNA damage response (DDR) is poorly understood. Here, we show that hyper-O-GlcNacylation leads to elevated insulin signaling, hyperproliferation, and DDR activation that mimic the glucose- and oxidative-stress-induced response. We discover a feedback mechanism involving key downstream effectors of DDR, ATM, ATR, and CHK1/2 that regulates OGT stability to promote O-GlcNAcylation and elevate DDR. This O-GlcNAc-dependent regulatory pathway is critical for maintaining gut homeostasis in Drosophila and the DDR in mouse embryonic stem cells (ESCs) and mouse embryonic fibroblasts (MEFs). Our findings reveal a conserved mechanistic link among O-GlcNAc cycling, stem cell self-renewal, and DDR with profound implications for stem-cell-derived diseases including cancer.


Asunto(s)
Daño del ADN/genética , Reparación del ADN/genética , N-Acetilglucosaminiltransferasas/metabolismo , Procesamiento Proteico-Postraduccional/genética , Células Madre/metabolismo , Animales , Homeostasis , Humanos , Ratones , Transducción de Señal
11.
Sci Rep ; 9(1): 12569, 2019 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-31467334

RESUMEN

Small numbers of hematopoietic stem cells (HSCs) balance self-renewal and differentiation to produce the diversity and abundance of cell types that make up the blood system. How nutrients are recruited to support this massive differentiation and proliferation process remains largely unknown. The unique metabolism of adult HSCs, which rely on glycolysis and glutaminolysis, suggests a potential role for the post-translational modification O-GlcNAc as a critical nutrient signal in these cells. Glutamine, glucose, and other metabolites drive the hexosamine biosynthetic pathway (HBP) ultimately leading to the O-GlcNAc modification of critical intracellular targets. Here, we used a conditional targeted genetic deletion of the enzyme that removes O-GlcNAc, O-GlcNAcase (OGA), to determine the consequences of blocked O-GlcNAc cycling on HSCs. Oga deletion in mouse HSCs resulted in greatly diminished progenitor pools, impaired stem cell self-renewal and nearly complete loss of competitive repopulation capacity. Further, early T cell specification was particularly sensitive to Oga deletion. Loss of Oga resulted in a doubling of apoptotic cells within the bone marrow and transcriptional deregulation of key genes involved in adult stem cell maintenance and lineage specification. These findings suggest that O-GlcNAc cycling plays a critical role in supporting HSC homeostasis and early thymocyte development.


Asunto(s)
Acetilglucosamina/metabolismo , Células Madre Hematopoyéticas/citología , Linfocitos T/citología , Animales , Apoptosis , Diferenciación Celular , Autorrenovación de las Células , Femenino , Eliminación de Gen , Masculino , Ratones , N-Acetilglucosaminiltransferasas/deficiencia , N-Acetilglucosaminiltransferasas/genética , Especificidad de Órganos , Timocitos/citología , Transcripción Genética
12.
FEBS Lett ; 592(23): 3943-3949, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29904918

RESUMEN

O-GlcNAcylation is an essential post-translational modification important for integrating metabolism with cell physiology. Using diverse model systems, studies of this evolutionarily conserved intracellular glycosylation have highlighted its role in stem cell maintenance, lineage specification, and disease. Although discovered over 30 years ago, the study of O-GlcNAc continues to evolve and uncover surprising roles for O-GlcNAc and the enzymes of O-GlcNAc cycling: O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA). In this review, using the immune system as a model of stem cell biology and cell fate determination, we discuss how O-GlcNAc is at the nexus of metabolism, proliferation, and disease.


Asunto(s)
Acetilglucosamina/metabolismo , Sistema Inmunológico/metabolismo , Procesamiento Proteico-Postraduccional , Linfocitos T/metabolismo , Diferenciación Celular , Glicosilación , Humanos , Modelos Biológicos , N-Acetilglucosaminiltransferasas/metabolismo , Células Madre/metabolismo
13.
Development ; 145(7)2018 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-29530881

RESUMEN

Imprinted genes are expressed from one parental allele and regulated by differential DNA methylation at imprinting control regions (ICRs). ICRs are reprogrammed in the germline through erasure and re-establishment of DNA methylation. Although much is known about DNA methylation establishment, DNA demethylation is less well understood. Recently, the Ten-Eleven Translocation proteins (TET1-3) have been shown to initiate DNA demethylation, with Tet1-/- mice exhibiting aberrant levels of imprinted gene expression and ICR methylation. Nevertheless, the role of TET1 in demethylating ICRs in the female germline and in controlling allele-specific expression remains unknown. Here, we examined ICR-specific DNA methylation in Tet1-/- germ cells and ascertained whether abnormal ICR methylation impacted imprinted gene expression in F1 hybrid somatic tissues derived from Tet1-/- eggs or sperm. We show that Tet1 deficiency is associated with hypermethylation of a subset of ICRs in germ cells. Moreover, ICRs with defective germline reprogramming exhibit aberrant DNA methylation and biallelic expression of linked imprinted genes in somatic tissues. Thus, we define a discrete set of genomic regions that require TET1 for germline reprogramming and discuss mechanisms for stochastic imprinting defects.


Asunto(s)
Metilación de ADN/genética , Proteínas de Unión al ADN/genética , Impresión Genómica/genética , Células Germinativas/metabolismo , Proteínas Proto-Oncogénicas/genética , Animales , Epigenómica , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa
14.
J Genomics ; 2: 77-88, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25031659

RESUMEN

It has long been recognized that men and women exhibit different risks for diverse disorders ranging from metabolic to autoimmune diseases. However, the underlying causes of these disparities remain obscure. Analysis of patients with chromosomal abnormalities, including Turner syndrome (45X) and Klinefelter syndrome (47XXY), has highlighted the importance of X-linked gene dosage as a contributing factor for disease susceptibility. Escape from X-inactivation and X-linked imprinting can result in transcriptional differences between normal men and women as well as in patients with sex chromosome abnormalities. Animal models support a role for X-linked gene dosage in disease with O-linked N-acetylglucosamine transferase (OGT) emerging as a prime candidate for a pleiotropic effector. OGT encodes a highly regulated nutrient-sensing epigenetic modifier with established links to immunity, metabolism and development.

15.
Biochem Biophys Res Commun ; 453(2): 201-7, 2014 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-24960196

RESUMEN

O-GlcNAcylation has emerged as a critical post-translational modification important for a wide array of cellular processes. This modification has been identified on a large pool of intracellular proteins that have wide-ranging roles, including transcriptional regulation, cell cycle progression, and signaling, among others. Interestingly, in mammals the single gene encoding O-GlcNAc Transferase (OGT) is located on the X-chromosome near the Xist locus suggesting that tight dosage regulation is necessary for normal development. Herein, we highlight the importance of OGT dosage and consider how its genomic location can contribute to a gender-specific increased risk for a number of diseases.


Asunto(s)
Genes Ligados a X , N-Acetilglucosaminiltransferasas/genética , Acetilglucosamina/metabolismo , Animales , Compensación de Dosificación (Genética) , Desarrollo Embrionario/genética , Desarrollo Embrionario/fisiología , Femenino , Impresión Genómica , Glicosilación , Humanos , Masculino , Ratones , Modelos Biológicos , N-Acetilglucosaminiltransferasas/metabolismo , Embarazo , Procesamiento Proteico-Postraduccional , Inactivación del Cromosoma X
16.
Bioconjug Chem ; 25(6): 1025-30, 2014 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-24866374

RESUMEN

The dynamic glycosylation of serine/threonine residues on nucleocytoplasmic proteins with a single N-acetylglucosamine (O-GlcNAcylation) is critical for many important cellular processes. Cellular O-GlcNAc levels are highly regulated by two enzymes: O-GlcNAc transferase (OGT) is responsible for GlcNAc addition and O-GlcNAcase (OGA) is responsible for removal of the sugar. The lack of a rapid and simple method for monitoring OGT activity has impeded the efficient discovery of potent OGT inhibitors. In this study we describe a novel, single-well OGT enzyme assay that utilizes 6 × His-tagged substrates, a chemoselective chemical reaction, and unpurified OGT. The high-throughput Ni-NTA Plate OGT Assay will facilitate discovery of potent OGT-specific inhibitors on versatile substrates and the characterization of new enzyme variants.


Asunto(s)
Pruebas de Enzimas , Inhibidores Enzimáticos/farmacología , N-Acetilglucosaminiltransferasas/antagonistas & inhibidores , N-Acetilglucosaminiltransferasas/metabolismo , Compuestos Organometálicos/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Humanos , Estructura Molecular , Níquel/química , Compuestos Organometálicos/química , Relación Estructura-Actividad , Especificidad por Sustrato/efectos de los fármacos , beta-N-Acetilhexosaminidasas/antagonistas & inhibidores , beta-N-Acetilhexosaminidasas/metabolismo
17.
Hum Reprod ; 27(5): 1249-59, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22416011

RESUMEN

BACKGROUND: Approximately 80% of childhood cancers can now be cured but a side effect of treatment results in about one-third of the surviving boys being infertile or severely subfertile when they reach reproductive age. Currently, more than 1 in 5000 men of reproductive age who are childhood cancer survivors suffer from this serious quality of life problem. It is possible to obtain a testicular biopsy before treatment to preserve the spermatogonial stem cells (SSCs) of the male by cryopreservation, but the results of long-term storage of SSCs on their subsequent functional ability to generate normal offspring has not been examined in any mammalian species. Moreover, it will be necessary to increase the number of these cryopreserved SSCs to remove any contaminating malignant cells and assure regeneration of spermatogenesis. METHODS AND RESULTS: In this report, we demonstrate that long-term cryopreservation (>14 years) of testis cells from mouse, rat, rabbit and baboon safeguards SSC viability, and that these cells can colonize the seminiferous tubules of recipient testes. Moreover, mouse and rat SSCs can be cultured and re-establish complete spermatogenesis, and fertile mouse progeny without apparent genetic or epigenetic errors were generated by the sperm produced. CONCLUSIONS: These findings provide a platform for fertility preservation in prepubertal boys undergoing gonadotoxic treatments.


Asunto(s)
Criopreservación , Preservación de la Fertilidad/métodos , Espermatogonias/citología , Células Madre , Animales , Femenino , Fertilidad , Masculino , Ratones , Papio/fisiología , Conejos , Ratas , Inyecciones de Esperma Intracitoplasmáticas , Espermatogonias/trasplante , Testículo/citología , Factores de Tiempo
18.
Curr Opin Genet Dev ; 22(2): 72-8, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22195775

RESUMEN

Genomic imprinting is an epigenetic process resulting in the monoallelic parent-of-origin-specific expression of a subset of genes in the mammalian genome. The parental alleles are differentially marked by DNA methylation during gametogenesis when the genomes are in separate compartments. How methylation machinery recognizes and differentially modifies these imprinted regions in germ cells remains a key question in the field. While studies have focused on determining a sequence signature that alone could distinguish imprinted regions from the rest of the genome, recent reports do not support such a hypothesis. Rather, it is becoming clear that features such as transcription, histone modifications and higher order chromatin are employed either individually or in combination to set up parental imprints.


Asunto(s)
Impresión Genómica , Animales , Metilación de ADN , Sitios Genéticos , Histonas/metabolismo , Humanos , Transcripción Genética
19.
Cell ; 146(1): 67-79, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21722948

RESUMEN

DNA methylation is a major epigenetic mechanism for gene silencing. Whereas methyltransferases mediate cytosine methylation, it is less clear how unmethylated regions in mammalian genomes are protected from de novo methylation and whether an active demethylating activity is involved. Here, we show that either knockout or catalytic inactivation of the DNA repair enzyme thymine DNA glycosylase (TDG) leads to embryonic lethality in mice. TDG is necessary for recruiting p300 to retinoic acid (RA)-regulated promoters, protection of CpG islands from hypermethylation, and active demethylation of tissue-specific developmentally and hormonally regulated promoters and enhancers. TDG interacts with the deaminase AID and the damage response protein GADD45a. These findings highlight a dual role for TDG in promoting proper epigenetic states during development and suggest a two-step mechanism for DNA demethylation in mammals, whereby 5-methylcytosine and 5-hydroxymethylcytosine are first deaminated by AID to thymine and 5-hydroxymethyluracil, respectively, followed by TDG-mediated thymine and 5-hydroxymethyluracil excision repair.


Asunto(s)
Metilación de ADN , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Timina ADN Glicosilasa/metabolismo , 5-Metilcitosina/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Citidina Desaminasa/metabolismo , Citosina/análogos & derivados , Citosina/metabolismo , Femenino , Técnicas de Sustitución del Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Timina ADN Glicosilasa/genética , Transcripción Genética
20.
Dev Biol ; 355(2): 349-57, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21600199

RESUMEN

Expression of coregulated imprinted genes, H19 and Igf2, is monoallelic and parent-of-origin-dependent. Like most imprinted genes, H19 and Igf2 are regulated by a differentially methylated imprinting control region (ICR). CTCF binding sites and DNA methylation at the ICR have previously been identified as key cis-acting elements required for proper H19/Igf2 imprinting. Here, we use mouse models to elucidate further the mechanism of ICR-mediated gene regulation. We specifically address the question of whether sequences outside of CTCF sites at the ICR are required for paternal H19 repression. To this end, we generated two types of mutant ICRs in the mouse: (i) deletion of intervening sequence between CTCF sites (H19(ICR∆IVS)), which changes size and CpG content at the ICR; and (ii) CpG depletion outside of CTCF sites (H19(ICR-8nrCG)), which only changes CpG content at the ICR. Individually, both mutant alleles (H19(ICR∆IVS) and H19(ICR-8nrCG)) show loss of imprinted repression of paternal H19. Interestingly, this loss of repression does not coincide with a detectable change in methylation at the H19 ICR or promoter. Thus, neither intact CTCF sites nor hypermethylation at the ICR is sufficient for maintaining the fully repressed state of the paternal H19 allele. Our findings demonstrate, for the first time in vivo, that sequence outside of CTCF sites at the ICR is required in cis for ICR-mediated imprinted repression at the H19/Igf2 locus. In addition, these results strongly implicate a novel role of ICR size and CpG density in paternal H19 repression.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Impresión Genómica/fisiología , ARN no Traducido/metabolismo , Elementos Reguladores de la Transcripción/fisiología , Proteínas Represoras/metabolismo , Animales , Southern Blotting , Factor de Unión a CCCTC , Cruzamientos Genéticos , Metilación de ADN/genética , Cartilla de ADN/genética , Electroporación , Vectores Genéticos/genética , Impresión Genómica/genética , Patrón de Herencia/genética , Ratones , Ratones Endogámicos C57BL , Mutagénesis Sitio-Dirigida , ARN Largo no Codificante , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Eliminación de Secuencia/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...