Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 140(5): 678-91, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-20211137

RESUMEN

The incorporation of histone H3 variants has been implicated in the epigenetic memory of cellular state. Using genome editing with zinc-finger nucleases to tag endogenous H3.3, we report genome-wide profiles of H3 variants in mammalian embryonic stem cells and neuronal precursor cells. Genome-wide patterns of H3.3 are dependent on amino acid sequence and change with cellular differentiation at developmentally regulated loci. The H3.3 chaperone Hira is required for H3.3 enrichment at active and repressed genes. Strikingly, Hira is not essential for localization of H3.3 at telomeres and many transcription factor binding sites. Immunoaffinity purification and mass spectrometry reveal that the proteins Atrx and Daxx associate with H3.3 in a Hira-independent manner. Atrx is required for Hira-independent localization of H3.3 at telomeres and for the repression of telomeric RNA. Our data demonstrate that multiple and distinct factors are responsible for H3.3 localization at specific genomic locations in mammalian cells.


Asunto(s)
Histonas/análisis , Telómero/química , Animales , Sitios de Unión , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Madre Embrionarias/metabolismo , Genoma , Chaperonas de Histonas/genética , Chaperonas de Histonas/metabolismo , Histonas/genética , Histonas/metabolismo , Ratones , Ratones Endogámicos C57BL , Telómero/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Sitio de Iniciación de la Transcripción
2.
Mol Syst Biol ; 18(8): e10473, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35996956

RESUMEN

Neuronal stimulation induced by the brain-derived neurotrophic factor (BDNF) triggers gene expression, which is crucial for neuronal survival, differentiation, synaptic plasticity, memory formation, and neurocognitive health. However, its role in chromatin regulation is unclear. Here, using temporal profiling of chromatin accessibility and transcription in mouse primary cortical neurons upon either BDNF stimulation or depolarization (KCl), we identify features that define BDNF-specific chromatin-to-gene expression programs. Enhancer activation is an early event in the regulatory control of BDNF-treated neurons, where the bZIP motif-binding Fos protein pioneered chromatin opening and cooperated with co-regulatory transcription factors (Homeobox, EGRs, and CTCF) to induce transcription. Deleting cis-regulatory sequences affect BDNF-mediated Arc expression, a regulator of synaptic plasticity. BDNF-induced accessible regions are linked to preferential exon usage by neurodevelopmental disorder-related genes and the heritability of neuronal complex traits, which were validated in human iPSC-derived neurons. Thus, we provide a comprehensive view of BDNF-mediated genome regulatory features using comparative genomic approaches to dissect mammalian neuronal stimulation.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Cromatina , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Factor Neurotrófico Derivado del Encéfalo/farmacología , Cromatina/genética , Cromatina/metabolismo , Humanos , Mamíferos/genética , Ratones , Neuronas/metabolismo , Factores de Transcripción/metabolismo
3.
Mol Cell ; 59(1): 89-103, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26073541

RESUMEN

Histone modification and DNA methylation are associated with varying epigenetic "landscapes," but detailed mechanistic and functional links between the two remain unclear. Using the ATRX-DNMT3-DNMT3L (ADD) domain of the DNA methyltransferase Dnmt3a as a paradigm, we apply protein engineering to dissect the molecular interactions underlying the recruitment of this enzyme to specific regions of chromatin in mouse embryonic stem cells (ESCs). By rendering the ADD domain insensitive to histone modification, specifically H3K4 methylation or H3T3 phosphorylation, we demonstrate the consequence of dysregulated Dnmt3a binding and activity. Targeting of a Dnmt3a mutant to H3K4me3 promoters decreases gene expression in a subset of developmental genes and alters ESC differentiation, whereas aberrant binding of another mutant to H3T3ph during mitosis promotes chromosome instability. Our studies support the general view that histone modification "reading" and DNA methylation are closely coupled in mammalian cells, and suggest an avenue for the functional assessment of chromatin-associated proteins.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , Células Madre Embrionarias/citología , Histonas/genética , Ingeniería de Proteínas , Animales , Diferenciación Celular , ADN Helicasas/genética , Metilación de ADN , ADN Metiltransferasa 3A , Ratones , Ratones Endogámicos C57BL , Mitosis/genética , Proteínas Nucleares/genética , Fosforilación , Regiones Promotoras Genéticas , Estructura Terciaria de Proteína , Proteína Nuclear Ligada al Cromosoma X
4.
Nature ; 522(7555): 240-244, 2015 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-25938714

RESUMEN

Transposable elements comprise roughly 40% of mammalian genomes. They have an active role in genetic variation, adaptation and evolution through the duplication or deletion of genes or their regulatory elements, and transposable elements themselves can act as alternative promoters for nearby genes, resulting in non-canonical regulation of transcription. However, transposable element activity can lead to detrimental genome instability, and hosts have evolved mechanisms to silence transposable element mobility appropriately. Recent studies have demonstrated that a subset of transposable elements, endogenous retroviral elements (ERVs) containing long terminal repeats (LTRs), are silenced through trimethylation of histone H3 on lysine 9 (H3K9me3) by ESET (also known as SETDB1 or KMT1E) and a co-repressor complex containing KRAB-associated protein 1 (KAP1; also known as TRIM28) in mouse embryonic stem cells. Here we show that the replacement histone variant H3.3 is enriched at class I and class II ERVs, notably those of the early transposon (ETn)/MusD family and intracisternal A-type particles (IAPs). Deposition at a subset of these elements is dependent upon the H3.3 chaperone complex containing α-thalassaemia/mental retardation syndrome X-linked (ATRX) and death-domain-associated protein (DAXX). We demonstrate that recruitment of DAXX, H3.3 and KAP1 to ERVs is co-dependent and occurs upstream of ESET, linking H3.3 to ERV-associated H3K9me3. Importantly, H3K9me3 is reduced at ERVs upon H3.3 deletion, resulting in derepression and dysregulation of adjacent, endogenous genes, along with increased retrotransposition of IAPs. Our study identifies a unique heterochromatin state marked by the presence of both H3.3 and H3K9me3, and establishes an important role for H3.3 in control of ERV retrotransposition in embryonic stem cells.


Asunto(s)
Células Madre Embrionarias/virología , Retrovirus Endógenos/genética , Silenciador del Gen , Histonas/metabolismo , Animales , Proteínas Portadoras/metabolismo , Línea Celular , Proteínas Co-Represoras , ADN Helicasas/metabolismo , Inestabilidad Genómica , Heterocromatina/genética , Heterocromatina/metabolismo , Histonas/química , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Metilación , Ratones , Chaperonas Moleculares , Proteínas Nucleares/metabolismo , Proteína Nuclear Ligada al Cromosoma X
6.
Nat Rev Genet ; 15(4): 259-71, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24614311

RESUMEN

Despite a conserved role for histones as general DNA packaging agents, it is now clear that another key function of these proteins is to confer variations in chromatin structure to ensure dynamic patterns of transcriptional regulation in eukaryotes. The incorporation of histone variants is particularly important to this process. Recent knockdown and knockout studies in various cellular systems, as well as direct mutational evidence from human cancers, now suggest a crucial role for histone variant regulation in processes as diverse as differentiation and proliferation, meiosis and nuclear reprogramming. In this Review, we provide an overview of histone variants in the context of their unique functions during mammalian germ cell and embryonic development, and examine the consequences of aberrant histone variant regulation in human disease.


Asunto(s)
Histonas/fisiología , Neoplasias/genética , Secuencia de Aminoácidos , Animales , Sitios de Unión , Cromatina/metabolismo , Desarrollo Embrionario , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Inestabilidad Genómica , Células Germinativas/metabolismo , Histonas/química , Humanos , Mutación Missense , Neoplasias/metabolismo , Cigoto/metabolismo
7.
Proc Natl Acad Sci U S A ; 114(10): E1885-E1894, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28223506

RESUMEN

CpG, 5'-C-phosphate-G-3', islands (CGIs) have long been known for their association with enhancers, silencers, and promoters, and for their epigenetic signatures. They are maintained in embryonic stem cells (ESCs) in a poised but inactive state via the formation of bivalent chromatin containing both active and repressive marks. CGIs also occur within coding sequences, where their functional role has remained obscure. Intragenic CGIs (iCGIs) are largely absent from housekeeping genes, but they are found in all genes associated with organ development and cell lineage control. In this paper, we investigated the epigenetic status of iCGIs and found that they too reside in bivalent chromatin in ESCs. Cell type-specific DNA methylation of iCGIs in differentiated cells was linked to the loss of both the H3K4me3 and H3K27me3 marks, and disruption of physical interaction with promoter regions, resulting in transcriptional activation of key regulators of differentiation such as PAXs, HOXs, and WNTs. The differential epigenetic modification of iCGIs appears to be mediated by cell type-specific transcription factors distinct from those bound by promoter, and these transcription factors may be involved in the hypermethylation of iCGIs upon cell differentiation. iCGIs thus play a key role in the cell type-specific regulation of transcription.


Asunto(s)
Diferenciación Celular/genética , Islas de CpG/genética , Metilación de ADN/genética , Epigénesis Genética/genética , Linaje de la Célula/genética , Cromatina/genética , Células Madre Embrionarias/citología , Elementos de Facilitación Genéticos/genética , Regulación del Desarrollo de la Expresión Génica , Histonas/genética , Humanos , Regiones Promotoras Genéticas
8.
Nature ; 548(7665): E7-E9, 2017 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-28770850
9.
Proc Natl Acad Sci U S A ; 112(22): 6820-7, 2015 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-25538301

RESUMEN

ATRX (the alpha thalassemia/mental retardation syndrome X-linked protein) is a member of the switch2/sucrose nonfermentable2 (SWI2/SNF2) family of chromatin-remodeling proteins and primarily functions at heterochromatic loci via its recognition of "repressive" histone modifications [e.g., histone H3 lysine 9 tri-methylation (H3K9me3)]. Despite significant roles for ATRX during normal neural development, as well as its relationship to human disease, ATRX function in the central nervous system is not well understood. Here, we describe ATRX's ability to recognize an activity-dependent combinatorial histone modification, histone H3 lysine 9 tri-methylation/serine 10 phosphorylation (H3K9me3S10ph), in postmitotic neurons. In neurons, this "methyl/phos" switch occurs exclusively after periods of stimulation and is highly enriched at heterochromatic repeats associated with centromeres. Using a multifaceted approach, we reveal that H3K9me3S10ph-bound Atrx represses noncoding transcription of centromeric minor satellite sequences during instances of heightened activity. Our results indicate an essential interaction between ATRX and a previously uncharacterized histone modification in the central nervous system and suggest a potential role for abnormal repetitive element transcription in pathological states manifested by ATRX dysfunction.


Asunto(s)
ADN Helicasas/metabolismo , Metilación de ADN/fisiología , Histonas/metabolismo , Neuronas/metabolismo , Proteínas Nucleares/metabolismo , Secuencias Repetitivas de Ácidos Nucleicos/fisiología , Animales , Calorimetría , Inmunoprecipitación de Cromatina , Cristalización , Cartilla de ADN/genética , Drosophila , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Silenciador del Gen , Humanos , Procesamiento de Imagen Asistido por Computador , Ratones , Ratones Endogámicos C57BL , Embarazo , Proteína Nuclear Ligada al Cromosoma X
10.
Proc Natl Acad Sci U S A ; 111(20): 7325-30, 2014 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-24799717

RESUMEN

Mature oocyte cytoplasm can reprogram somatic cell nuclei to the pluripotent state through a series of sequential events including protein exchange between the donor nucleus and ooplasm, chromatin remodeling, and pluripotency gene reactivation. Maternal factors that are responsible for this reprogramming process remain largely unidentified. Here, we demonstrate that knockdown of histone variant H3.3 in mouse oocytes results in compromised reprogramming and down-regulation of key pluripotency genes; and this compromised reprogramming for developmental potentials and transcription of pluripotency genes can be rescued by injecting exogenous H3.3 mRNA, but not H3.2 mRNA, into oocytes in somatic cell nuclear transfer embryos. We show that maternal H3.3, and not H3.3 in the donor nucleus, is essential for successful reprogramming of somatic cell nucleus into the pluripotent state. Furthermore, H3.3 is involved in this reprogramming process by remodeling the donor nuclear chromatin through replacement of donor nucleus-derived H3 with de novo synthesized maternal H3.3 protein. Our study shows that H3.3 is a crucial maternal factor for oocyte reprogramming and provides a practical model to directly dissect the oocyte for its reprogramming capacity.


Asunto(s)
Núcleo Celular/metabolismo , Reprogramación Celular , Regulación del Desarrollo de la Expresión Génica , Histonas/química , Oocitos/citología , Animales , Cromatina/metabolismo , Citoplasma/metabolismo , Femenino , Ratones , Técnicas de Transferencia Nuclear , Oocitos/metabolismo , ARN Interferente Pequeño/metabolismo , Análisis de Secuencia de ARN
11.
Proc Natl Acad Sci U S A ; 109(16): E962-71, 2012 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-22371606

RESUMEN

Dysregulation of the transcriptional repressor element-1 silencing transcription factor (REST)/neuron-restrictive silencer factor is important in a broad range of diseases, including cancer, diabetes, and heart disease. The role of REST-dependent epigenetic modifications in neurodegeneration is less clear. Here, we show that neuronal insults trigger activation of REST and CoREST in a clinically relevant model of ischemic stroke and that REST binds a subset of "transcriptionally responsive" genes (gria2, grin1, chrnb2, nefh, nfκb2, trpv1, chrm4, and syt6), of which the AMPA receptor subunit GluA2 is a top hit. Genes with enriched REST exhibited decreased mRNA and protein. We further show that REST assembles with CoREST, mSin3A, histone deacetylases 1 and 2, histone methyl-transferase G9a, and methyl CpG binding protein 2 at the promoters of target genes, where it orchestrates epigenetic remodeling and gene silencing. RNAi-mediated depletion of REST or administration of dominant-negative REST delivered directly into the hippocampus in vivo prevents epigenetic modifications, restores gene expression, and rescues hippocampal neurons. These findings document a causal role for REST-dependent epigenetic remodeling in the neurodegeneration associated with ischemic stroke and identify unique therapeutic targets for the amelioration of hippocampal injury and cognitive deficits.


Asunto(s)
Epigénesis Genética/genética , Epigenómica , Neuronas/metabolismo , Proteínas Represoras/genética , Animales , Western Blotting , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/patología , Muerte Celular , Células Cultivadas , Proteínas Co-Represoras/genética , Proteínas Co-Represoras/metabolismo , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Isquemia/complicaciones , Masculino , Microscopía Fluorescente , Neuronas/patología , Regiones Promotoras Genéticas/genética , Unión Proteica , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Receptores AMPA/genética , Receptores AMPA/metabolismo , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/metabolismo
12.
Genesis ; 52(12): 959-66, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25262655

RESUMEN

Chromatin remodeling via incorporation of histone variants plays a key role in the regulation of embryonic development. The histone variant H3.3 has been associated with a number of early events including formation of the paternal pronucleus upon fertilization. The small number of amino acid differences between H3.3 and its canonical counterparts (H3.1 and H3.2) has limited studies of the developmental significance of H3.3 deposition into chromatin due to difficulties in distinguishing the H3 isoforms. To this end, we used zinc-finger nuclease (ZFN) mediated gene editing to introduce a small C-terminal hemagglutinin (HA) tag to the endogenous H3.3B locus in mouse embryonic stem cells (ESCs), along with an internal ribosome entry site (IRES) and a separately translated fluorescent reporter of expression. This system will allow detection of expression driven by the reporter in cells, animals, and embryos, and will facilitate investigation of differential roles of paternal and maternal H3.3 protein during embryogenesis that would not be possible using variant-specific antibodies. Further, the ability to monitor endogenous H3.3 protein in various cell lineages will enhance our understanding of the dynamics of this histone variant over the course of development.


Asunto(s)
Embrión de Mamíferos/metabolismo , Ingeniería Genética/métodos , Histonas/genética , Histonas/metabolismo , Animales , Cromatina/genética , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Desarrollo Embrionario , Femenino , Regulación del Desarrollo de la Expresión Génica , Sitios Genéticos , Variación Genética , Masculino , Ratones , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
13.
Life Sci Alliance ; 7(5)2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38418090

RESUMEN

During development, different tissues acquire distinct lipotypes that are coupled to tissue function and homeostasis. In the brain, where complex membrane trafficking systems are required for neural function, specific glycerophospholipids, sphingolipids, and cholesterol are highly abundant, and defective lipid metabolism is associated with abnormal neural development and neurodegenerative disease. Notably, the production of specific lipotypes requires appropriate programming of the underlying lipid metabolic machinery during development, but when and how this occurs is unclear. To address this, we used high-resolution MSALL lipidomics to generate an extensive time-resolved resource of mouse brain development covering early embryonic and postnatal stages. This revealed a distinct bifurcation in the establishment of the neural lipotype, whereby the canonical lipid biomarkers 22:6-glycerophospholipids and 18:0-sphingolipids begin to be produced in utero, whereas cholesterol attains its characteristic high levels after birth. Using the resource as a reference, we next examined to which extent this can be recapitulated by commonly used protocols for in vitro neuronal differentiation of stem cells. Here, we found that the programming of the lipid metabolic machinery is incomplete and that stem cell-derived cells can only partially acquire a neural lipotype when the cell culture media is supplemented with brain-specific lipid precursors. Altogether, our work provides an extensive lipidomic resource for early mouse brain development and highlights a potential caveat when using stem cell-derived neuronal progenitors for mechanistic studies of lipid biochemistry, membrane biology and biophysics, which nonetheless can be mitigated by further optimizing in vitro differentiation protocols.


Asunto(s)
Enfermedades Neurodegenerativas , Ratones , Animales , Células Madre/metabolismo , Neuronas/metabolismo , Esfingolípidos/metabolismo , Colesterol , Glicerofosfolípidos/metabolismo
14.
Proc Natl Acad Sci U S A ; 107(32): 14075-80, 2010 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-20651253

RESUMEN

The histone variant H3.3 is implicated in the formation and maintenance of specialized chromatin structure in metazoan cells. H3.3-containing nucleosomes are assembled in a replication-independent manner by means of dedicated chaperone proteins. We previously identified the death domain associated protein (Daxx) and the alpha-thalassemia X-linked mental retardation protein (ATRX) as H3.3-associated proteins. Here, we report that the highly conserved N terminus of Daxx interacts directly with variant-specific residues in the H3.3 core. Recombinant Daxx assembles H3.3/H4 tetramers on DNA templates, and the ATRX-Daxx complex catalyzes the deposition and remodeling of H3.3-containing nucleosomes. We find that the ATRX-Daxx complex is bound to telomeric chromatin, and that both components of this complex are required for H3.3 deposition at telomeres in murine embryonic stem cells (ESCs). These data demonstrate that Daxx functions as an H3.3-specific chaperone and facilitates the deposition of H3.3 at heterochromatin loci in the context of the ATRX-Daxx complex.


Asunto(s)
Proteínas Portadoras/fisiología , Ensamble y Desensamble de Cromatina , ADN Helicasas/metabolismo , Chaperonas de Histonas/metabolismo , Histonas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas Nucleares/metabolismo , Proteínas Nucleares/fisiología , Telómero , Animales , Proteínas Portadoras/metabolismo , Proteínas Co-Represoras , Células Madre Embrionarias , Heterocromatina , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Chaperonas Moleculares , Complejos Multiproteicos , Nucleosomas/metabolismo , Unión Proteica , Proteína Nuclear Ligada al Cromosoma X
15.
Science ; 379(6636): 1010-1015, 2023 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-36893247

RESUMEN

Dynamic measurements of molecular machines can provide invaluable insights into their mechanism, but these measurements have been challenging in living cells. Here, we developed live-cell tracking of single fluorophores with nanometer spatial and millisecond temporal resolution in two and three dimensions using the recently introduced super-resolution technique MINFLUX. Using this approach, we resolved the precise stepping motion of the motor protein kinesin-1 as it walked on microtubules in living cells. Nanoscopic tracking of motors walking on the microtubules of fixed cells also enabled us to resolve the architecture of the microtubule cytoskeleton with protofilament resolution.


Asunto(s)
Células , Cinesinas , Microscopía Fluorescente , Microtúbulos , Células/química , Células/metabolismo , Colorantes Fluorescentes/análisis , Cinesinas/química , Cinesinas/metabolismo , Microscopía Fluorescente/instrumentación , Microscopía Fluorescente/métodos , Microtúbulos/química , Microtúbulos/metabolismo , Movimiento (Física) , Humanos
16.
Mol Omics ; 18(4): 296-314, 2022 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-35044400

RESUMEN

Histone variants, such as histone H3.3, replace canonical histones within the nucleosome to alter chromatin accessibility and gene expression. Although the biological roles of selected histone post-translational modifications (PTMs) have been extensively characterized, the potential differences in the function of a given PTM on different histone variants is almost always elusive. By applying proteomics and genomics techniques, we investigate the role of lysine 27 tri-methylation specifically on the histone variant H3.3 (H3.3K27me3) in the context of mouse embryonic stem cell pluripotency and differentiation as a model system for development. We demonstrate that while the steady state overall levels of methylation on both H3K27 and H3.3K27 decrease during differentiation, methylation dynamics studies indicate that methylation on H3.3K27 is maintained more than on H3K27. Using a custom-made antibody, we identify a unique enrichment of H3.3K27me3 at lineage-specific genes, such as olfactory receptor genes, and at binding motifs for the transcription factors FOXJ2/3. REST, a predicted FOXJ2/3 target that acts as a transcriptional repressor of terminal neuronal genes, was identified with H3.3K27me3 at its promoter region. H3.3K27A mutant cells confirmed an upregulation of FOXJ2/3 targets upon the loss of methylation at H3.3K27. Thus, while canonical H3K27me3 has been characterized to regulate the expression of transcription factors that play a general role in differentiation, our work suggests H3.3K27me3 is essential for regulating distinct terminal differentiation genes. This work highlights the importance of understanding the effects of PTMs not only on canonical histones but also on specific histone variants, as they may exhibit distinct roles.


Asunto(s)
Histonas , Lisina , Animales , Diferenciación Celular/genética , Histonas/genética , Histonas/metabolismo , Lisina/química , Metilación , Ratones , Procesamiento Proteico-Postraduccional , Factores de Transcripción/genética
17.
Nat Biotechnol ; 40(3): 382-390, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34663920

RESUMEN

Phosphorylation is a critical post-translational modification involved in the regulation of almost all cellular processes. However, fewer than 5% of thousands of recently discovered phosphosites have been functionally annotated. In this study, we devised a chemical genetic approach to study the functional relevance of phosphosites in Saccharomyces cerevisiae. We generated 474 yeast strains with mutations in specific phosphosites that were screened for fitness in 102 conditions, along with a gene deletion library. Of these phosphosites, 42% exhibited growth phenotypes, suggesting that these are more likely functional. We inferred their function based on the similarity of their growth profiles with that of gene deletions and validated a subset by thermal proteome profiling and lipidomics. A high fraction exhibited phenotypes not seen in the corresponding gene deletion, suggestive of a gain-of-function effect. For phosphosites conserved in humans, the severity of the yeast phenotypes is indicative of their human functional relevance. This high-throughput approach allows for functionally characterizing individual phosphosites at scale.


Asunto(s)
Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Fosforilación , Procesamiento Proteico-Postraduccional/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
18.
Proc Natl Acad Sci U S A ; 105(12): 4892-7, 2008 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-18347331

RESUMEN

Transient forebrain or global ischemia induces delayed neuronal death in vulnerable CA1 pyramidal cells with many features of apoptosis. A brief period of ischemia, i.e., ischemic preconditioning, affords robust protection of CA1 neurons against a subsequent more prolonged ischemic challenge. Here we show that preconditioning acts via PI3K/Akt signaling to block the ischemia-induced cascade involving mitochondrial translocation of Bad, assembly of Bad with Bcl-x(L), cleavage of Bcl-x(L) to form its prodeath fragment, DeltaN-Bcl-x(L), activation of large-conductance channels in the mitochondrial outer membrane, mitochondrial release of cytochrome c and Smac/DIABLO (second mitochondria-derived activator of caspases/direct IAP-binding protein with low pI), caspase activation, and neuronal death. These findings show how preconditioning acts to prevent the release of cytochrome c and Smac/DIABLO from mitochondria and to preserve the integrity of the mitochondrial membrane. The specific PI3K inhibitor LY294002 administered in vivo 1 h before or immediately after ischemia or up to 120 h later significantly reverses preconditioning-induced protection, indicating a requirement for sustained PI3K signaling in ischemic tolerance. These findings implicate PI3K/Akt signaling in maintenance of the integrity of the mitochondrial outer membrane.


Asunto(s)
Hipocampo/metabolismo , Precondicionamiento Isquémico , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Mitocondrias/metabolismo , Neuronas/metabolismo , Proteína Letal Asociada a bcl/metabolismo , Proteína bcl-X/metabolismo , Animales , Apoptosis/efectos de los fármacos , Isquemia Encefálica/enzimología , Inhibidores de Caspasas , Cromonas/farmacología , Hipocampo/citología , Hipocampo/efectos de los fármacos , Hipocampo/enzimología , Activación del Canal Iónico/efectos de los fármacos , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Morfolinas/farmacología , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/enzimología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
19.
Sci Rep ; 11(1): 10266, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33986420

RESUMEN

Antarctic marine biological variability modulates climate systems via the biological pump. However, the knowledge of biological response in the Southern Ocean to climate variability still has been lack of understanding owing to limited ocean color data in the high latitude region. We investigated the surface chlorophyll concentration responses to the Southern annular mode (SAM) in the marginal sea of the Southern ocean using satellite observation and reanalysis data focusing on the austral summer. The positive phase of SAM is associated with enhanced and poleward-shifted westerly winds, leading to physical and biogeochemical responses over the Southern ocean. Our result indicates that chlorophyll has strong zonally asymmetric responses to SAM owing to different limiting factors of phytoplankton growth per region. For the positive SAM phase, chlorophyll tends to increase in the western Amundsen-Ross Sea but decreases in the D'Urville Sea. It is suggested that the distinct limiting factors are associated with the seasonal variability of sea ice and upwelling per region.

20.
Cell Stem Cell ; 28(7): 1291-1306.e10, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33848472

RESUMEN

Generation of induced oligodendrocyte progenitor cells (iOPCs) from somatic fibroblasts is a strategy for cell-based therapy of myelin diseases. However, iOPC generation is inefficient, and the resulting iOPCs exhibit limited expansion and differentiation competence. Here we overcome these limitations by transducing an optimized transcription factor combination into a permissive donor phenotype, the pericyte. Pericyte-derived iOPCs (PC-iOPCs) are stably expandable and functionally myelinogenic with high differentiation competence. Unexpectedly, however, we found that PC-iOPCs are metastable so that they can produce myelination-competent oligodendrocytes or revert to their original identity in a context-dependent fashion. Phenotypic reversion of PC-iOPCs is tightly linked to memory of their original transcriptome and epigenome. Phenotypic reversion can be disconnected from this donor cell memory effect, and in vivo myelination can eventually be achieved by transplantation of O4+ pre-oligodendrocytes. Our data show that donor cell source and memory can contribute to the fate and stability of directly converted cells.


Asunto(s)
Vaina de Mielina , Oligodendroglía , Diferenciación Celular , Fibroblastos , Células Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA