Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20.455
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Immunol ; 40: 387-411, 2022 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-35119910

RESUMEN

Cell identity and function largely rely on the programming of transcriptomes during development and differentiation. Signature gene expression programs are orchestrated by regulatory circuits consisting of cis-acting promoters and enhancers, which respond to a plethora of cues via the action of transcription factors. In turn, transcription factors direct epigenetic modifications to revise chromatin landscapes, and drive contacts between distal promoter-enhancer combinations. In immune cells, regulatory circuits for effector genes are especially complex and flexible, utilizing distinct sets of transcription factors and enhancers, depending on the cues each cell type receives during an infection, after sensing cellular damage, or upon encountering a tumor. Here, we review major players in the coordination of gene regulatory programs within innate and adaptive immune cells, as well as integrative omics approaches that can be leveraged to decipher their underlying circuitry.


Asunto(s)
Cromatina , Redes Reguladoras de Genes , Animales , Regulación de la Expresión Génica , Humanos , Regiones Promotoras Genéticas , Factores de Transcripción/genética
2.
Annu Rev Immunol ; 39: 667-693, 2021 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-33637018

RESUMEN

Traditionally, the innate and adaptive immune systems are differentiated by their specificity and memory capacity. In recent years, however, this paradigm has shifted: Cells of the innate immune system appear to be able to gain memory characteristics after transient stimulation, resulting in an enhanced response upon secondary challenge. This phenomenon has been called trained immunity. Trained immunity is characterized by nonspecific increased responsiveness, mediated via extensive metabolic and epigenetic reprogramming. Trained immunity explains the heterologous effects of vaccines, which result in increased protection against secondary infections. However, in chronic inflammatory conditions, trained immunity can induce maladaptive effects and contribute to hyperinflammation and progression of cardiovascular disease, autoinflammatory syndromes, and neuroinflammation. In this review we summarize the current state of the field of trained immunity, its mechanisms, and its roles in both health and disease.


Asunto(s)
Memoria Inmunológica , Vacunas , Animales , Diferenciación Celular , Humanos , Sistema Inmunológico , Inmunidad Innata
3.
Annu Rev Immunol ; 39: 395-416, 2021 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-33902315

RESUMEN

Recent evidence supports the notion that mitochondrial metabolism is necessary for T cell activation, proliferation, and function. Mitochondrial metabolism supports T cell anabolism by providing key metabolites for macromolecule synthesis and generating metabolites for T cell function. In this review, we focus on how mitochondrial metabolism controls conventional and regulatory T cell fates and function.


Asunto(s)
Inmunidad Celular , Mitocondrias , Animales , Humanos
4.
Annu Rev Immunol ; 36: 221-246, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29328786

RESUMEN

Researchers are intensifying efforts to understand the mechanisms by which changes in metabolic states influence differentiation programs. An emerging objective is to define how fluctuations in metabolites influence the epigenetic states that contribute to differentiation programs. This is because metabolites such as S-adenosylmethionine, acetyl-CoA, α-ketoglutarate, 2-hydroxyglutarate, and butyrate are donors, substrates, cofactors, and antagonists for the activities of epigenetic-modifying complexes and for epigenetic modifications. We discuss this topic from the perspective of specialized CD4+ T cells as well as effector and memory T cell differentiation programs. We also highlight findings from embryonic stem cells that give mechanistic insight into how nutrients processed through pathways such as glycolysis, glutaminolysis, and one-carbon metabolism regulate metabolite levels to influence epigenetic events and discuss similar mechanistic principles in T cells. Finally, we highlight how dysregulated environments, such as the tumor microenvironment, might alter programming events.


Asunto(s)
Diferenciación Celular/genética , Diferenciación Celular/inmunología , Metabolismo Energético , Epigénesis Genética , Animales , Biomarcadores , Regulación del Desarrollo de la Expresión Génica , Humanos , Neoplasias/etiología , Neoplasias/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
5.
Annu Rev Immunol ; 35: 1-30, 2017 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-27912315

RESUMEN

Genome technologies have defined a complex genetic architecture in major infectious, inflammatory, and autoimmune disorders. High density marker arrays and Immunochips have powered genome-wide association studies (GWAS) that have mapped nearly 450 genetic risk loci in 22 major inflammatory diseases, including a core of common genes that play a central role in pathological inflammation. Whole-exome and whole-genome sequencing have identified more than 265 genes in which mutations cause primary immunodeficiencies and rare forms of severe inflammatory bowel disease. Combined analysis of inflammatory disease GWAS and primary immunodeficiencies point to shared proteins and pathways that are required for immune cell development and protection against infections and are also associated with pathological inflammation. Finally, sequencing of chromatin immunoprecipitates containing specific transcription factors, with parallel RNA sequencing, has charted epigenetic regulation of gene expression by proinflammatory transcription factors in immune cells, providing complementary information to characterize morbid genes at infectious and inflammatory disease loci.


Asunto(s)
Enfermedades Autoinmunes/genética , Síndromes de Inmunodeficiencia/genética , Infecciones/genética , Inflamación/genética , Vacunas/inmunología , Animales , Epigénesis Genética , Exoma/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunidad/genética , Infecciones/inmunología , Riesgo
6.
Annu Rev Immunol ; 35: 53-84, 2017 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-27912316

RESUMEN

Helper T (Th) cell subsets direct immune responses by producing signature cytokines. Th2 cells produce IL-4, IL-5, and IL-13, which are important in humoral immunity and protection from helminth infection and are central to the pathogenesis of many allergic inflammatory diseases. Molecular analysis of Th2 cell differentiation and maintenance of function has led to recent discoveries that have refined our understanding of Th2 cell biology. Epigenetic regulation of Gata3 expression by chromatin remodeling complexes such as Polycomb and Trithorax is crucial for maintaining Th2 cell identity. In the context of allergic diseases, memory-type pathogenic Th2 cells have been identified in both mice and humans. To better understand these disease-driving cell populations, we have developed a model called the pathogenic Th population disease induction model. The concept of defined subsets of pathogenic Th cells may spur new, effective strategies for treating intractable chronic inflammatory disorders.


Asunto(s)
Helmintiasis/inmunología , Hipersensibilidad/inmunología , Células Th2/inmunología , Animales , Diferenciación Celular , Modelos Animales de Enfermedad , Epigénesis Genética , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Inmunidad Humoral , Memoria Inmunológica , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Interleucina-5/metabolismo , Ratones , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo
7.
Cell ; 187(15): 4030-4042.e13, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-38908367

RESUMEN

Insufficient telomerase activity, stemming from low telomerase reverse transcriptase (TERT) gene transcription, contributes to telomere dysfunction and aging pathologies. Besides its traditional function in telomere synthesis, TERT acts as a transcriptional co-regulator of genes pivotal in aging and age-associated diseases. Here, we report the identification of a TERT activator compound (TAC) that upregulates TERT transcription via the MEK/ERK/AP-1 cascade. In primary human cells and naturally aged mice, TAC-induced elevation of TERT levels promotes telomere synthesis, blunts tissue aging hallmarks with reduced cellular senescence and inflammatory cytokines, and silences p16INK4a expression via upregulation of DNMT3B-mediated promoter hypermethylation. In the brain, TAC alleviates neuroinflammation, increases neurotrophic factors, stimulates adult neurogenesis, and preserves cognitive function without evident toxicity, including cancer risk. Together, these findings underscore TERT's critical role in aging processes and provide preclinical proof of concept for physiological TERT activation as a strategy to mitigate multiple aging hallmarks and associated pathologies.


Asunto(s)
Envejecimiento , Metilación de ADN , Telomerasa , Telomerasa/metabolismo , Telomerasa/genética , Humanos , Animales , Ratones , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , Senescencia Celular , Regiones Promotoras Genéticas , ADN Metiltransferasa 3B , Encéfalo/metabolismo , Telómero/metabolismo , Ratones Endogámicos C57BL , Masculino , Factor de Transcripción AP-1/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Neurogénesis
8.
Cell ; 187(13): 3338-3356.e30, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38810644

RESUMEN

Suspended animation states allow organisms to survive extreme environments. The African turquoise killifish has evolved diapause as a form of suspended development to survive a complete drought. However, the mechanisms underlying the evolution of extreme survival states are unknown. To understand diapause evolution, we performed integrative multi-omics (gene expression, chromatin accessibility, and lipidomics) in the embryos of multiple killifish species. We find that diapause evolved by a recent remodeling of regulatory elements at very ancient gene duplicates (paralogs) present in all vertebrates. CRISPR-Cas9-based perturbations identify the transcription factors REST/NRSF and FOXOs as critical for the diapause gene expression program, including genes involved in lipid metabolism. Indeed, diapause shows a distinct lipid profile, with an increase in triglycerides with very-long-chain fatty acids. Our work suggests a mechanism for the evolution of complex adaptations and offers strategies to promote long-term survival by activating suspended animation programs in other species.


Asunto(s)
Diapausa , Animales , Evolución Biológica , Diapausa/genética , Embrión no Mamífero/metabolismo , Fundulidae/genética , Fundulidae/metabolismo , Regulación del Desarrollo de la Expresión Génica , Peces Killi/genética , Peces Killi/metabolismo , Metabolismo de los Lípidos/genética , Proteínas de Peces/genética , Masculino , Femenino
9.
Cell ; 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39243764

RESUMEN

There is documented sex disparity in cutaneous melanoma incidence and mortality, increasing disproportionately with age and in the male sex. However, the underlying mechanisms remain unclear. While biological sex differences and inherent immune response variability have been assessed in tumor cells, the role of the tumor-surrounding microenvironment, contextually in aging, has been overlooked. Here, we show that skin fibroblasts undergo age-mediated, sex-dependent changes in their proliferation, senescence, ROS levels, and stress response. We find that aged male fibroblasts selectively drive an invasive, therapy-resistant phenotype in melanoma cells and promote metastasis in aged male mice by increasing AXL expression. Intrinsic aging in male fibroblasts mediated by EZH2 decline increases BMP2 secretion, which in turn drives the slower-cycling, highly invasive, and therapy-resistant melanoma cell phenotype, characteristic of the aged male TME. Inhibition of BMP2 activity blocks the emergence of invasive phenotypes and sensitizes melanoma cells to BRAF/MEK inhibition.

10.
Cell ; 187(18): 5010-5028.e24, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39094570

RESUMEN

Faithful transfer of parental histones to newly replicated daughter DNA strands is critical for inheritance of epigenetic states. Although replication proteins that facilitate parental histone transfer have been identified, how intact histone H3-H4 tetramers travel from the front to the back of the replication fork remains unknown. Here, we use AlphaFold-Multimer structural predictions combined with biochemical and genetic approaches to identify the Mrc1/CLASPIN subunit of the replisome as a histone chaperone. Mrc1 contains a conserved histone-binding domain that forms a brace around the H3-H4 tetramer mimicking nucleosomal DNA and H2A-H2B histones, is required for heterochromatin inheritance, and promotes parental histone recycling during replication. We further identify binding sites for the FACT histone chaperone in Swi1/TIMELESS and DNA polymerase α that are required for heterochromatin inheritance. We propose that Mrc1, in concert with FACT acting as a mobile co-chaperone, coordinates the distribution of parental histones to newly replicated DNA.


Asunto(s)
Replicación del ADN , Epigénesis Genética , Heterocromatina , Histonas , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Histonas/metabolismo , Heterocromatina/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas del Grupo de Alta Movilidad/genética , Factores de Elongación Transcripcional/metabolismo , Factores de Elongación Transcripcional/genética , Chaperonas de Histonas/metabolismo , Chaperonas Moleculares/metabolismo , ADN Polimerasa I/metabolismo , ADN Polimerasa I/genética
11.
Cell ; 187(5): 1238-1254.e14, 2024 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-38367616

RESUMEN

CD4+ T cells with latent HIV-1 infection persist despite treatment with antiretroviral agents and represent the main barrier to a cure of HIV-1 infection. Pharmacological disruption of viral latency may expose HIV-1-infected cells to host immune activity, but the clinical efficacy of latency-reversing agents for reducing HIV-1 persistence remains to be proven. Here, we show in a randomized-controlled human clinical trial that the histone deacetylase inhibitor panobinostat, when administered in combination with pegylated interferon-α2a, induces a structural transformation of the HIV-1 reservoir cell pool, characterized by a disproportionate overrepresentation of HIV-1 proviruses integrated in ZNF genes and in chromatin regions with reduced H3K27ac marks, the molecular target sites for panobinostat. By contrast, proviruses near H3K27ac marks were actively selected against, likely due to increased susceptibility to panobinostat. These data suggest that latency-reversing treatment can increase the immunological vulnerability of HIV-1 reservoir cells and accelerate the selection of epigenetically privileged HIV-1 proviruses.


Asunto(s)
Infecciones por VIH , VIH-1 , Inhibidores de Histona Desacetilasas , Interferón-alfa , Panobinostat , Provirus , Humanos , Infecciones por VIH/tratamiento farmacológico , VIH-1/genética , Panobinostat/uso terapéutico , Provirus/efectos de los fármacos , Latencia del Virus , Inhibidores de Histona Desacetilasas/uso terapéutico , Interferón-alfa/uso terapéutico
12.
Cell ; 186(12): 2531-2543.e11, 2023 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-37295401

RESUMEN

RNA editing is a widespread epigenetic process that can alter the amino acid sequence of proteins, termed "recoding." In cephalopods, most transcripts are recoded, and recoding is hypothesized to be an adaptive strategy to generate phenotypic plasticity. However, how animals use RNA recoding dynamically is largely unexplored. We investigated the function of cephalopod RNA recoding in the microtubule motor proteins kinesin and dynein. We found that squid rapidly employ RNA recoding in response to changes in ocean temperature, and kinesin variants generated in cold seawater displayed enhanced motile properties in single-molecule experiments conducted in the cold. We also identified tissue-specific recoded squid kinesin variants that displayed distinct motile properties. Finally, we showed that cephalopod recoding sites can guide the discovery of functional substitutions in non-cephalopod kinesin and dynein. Thus, RNA recoding is a dynamic mechanism that generates phenotypic plasticity in cephalopods and can inform the characterization of conserved non-cephalopod proteins.


Asunto(s)
Cefalópodos , Dineínas , Animales , Dineínas/genética , Dineínas/metabolismo , Cinesinas/genética , Cinesinas/metabolismo , ARN/metabolismo , Cefalópodos/genética , Cefalópodos/metabolismo , Proteínas/metabolismo , Microtúbulos/metabolismo , Proteínas de Microtúbulos , Miosinas/metabolismo
13.
Cell ; 186(5): 957-974.e28, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36812912

RESUMEN

Bats are distinctive among mammals due to their ability to fly, use laryngeal echolocation, and tolerate viruses. However, there are currently no reliable cellular models for studying bat biology or their response to viral infections. Here, we created induced pluripotent stem cells (iPSCs) from two species of bats: the wild greater horseshoe bat (Rhinolophus ferrumequinum) and the greater mouse-eared bat (Myotis myotis). The iPSCs from both bat species showed similar characteristics and had a gene expression profile resembling that of cells attacked by viruses. They also had a high number of endogenous viral sequences, particularly retroviruses. These results suggest that bats have evolved mechanisms to tolerate a large load of viral sequences and may have a more intertwined relationship with viruses than previously thought. Further study of bat iPSCs and their differentiated progeny will provide insights into bat biology, virus host relationships, and the molecular basis of bats' special traits.


Asunto(s)
Quirópteros , Células Madre Pluripotentes , Virosis , Virus , Animales , Virus/genética , Transcriptoma , Filogenia
14.
Cell ; 186(26): 5840-5858.e36, 2023 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-38134876

RESUMEN

Short tandem repeat (STR) instability causes transcriptional silencing in several repeat expansion disorders. In fragile X syndrome (FXS), mutation-length expansion of a CGG STR represses FMR1 via local DNA methylation. Here, we find megabase-scale H3K9me3 domains on autosomes and encompassing FMR1 on the X chromosome in FXS patient-derived iPSCs, iPSC-derived neural progenitors, EBV-transformed lymphoblasts, and brain tissue with mutation-length CGG expansion. H3K9me3 domains connect via inter-chromosomal interactions and demarcate severe misfolding of TADs and loops. They harbor long synaptic genes replicating at the end of S phase, replication-stress-induced double-strand breaks, and STRs prone to stepwise somatic instability. CRISPR engineering of the mutation-length CGG to premutation length reverses H3K9me3 on the X chromosome and multiple autosomes, refolds TADs, and restores gene expression. H3K9me3 domains can also arise in normal-length iPSCs created with perturbations linked to genome instability, suggesting their relevance beyond FXS. Our results reveal Mb-scale heterochromatinization and trans interactions among loci susceptible to instability.


Asunto(s)
Síndrome del Cromosoma X Frágil , Humanos , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/metabolismo , Expansión de Repetición de Trinucleótido , Metilación de ADN , Mutación , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo
15.
Cell ; 185(25): 4841-4860.e25, 2022 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-36493756

RESUMEN

We present a multiomic cell atlas of human lung development that combines single-cell RNA and ATAC sequencing, high-throughput spatial transcriptomics, and single-cell imaging. Coupling single-cell methods with spatial analysis has allowed a comprehensive cellular survey of the epithelial, mesenchymal, endothelial, and erythrocyte/leukocyte compartments from 5-22 post-conception weeks. We identify previously uncharacterized cell states in all compartments. These include developmental-specific secretory progenitors and a subtype of neuroendocrine cell related to human small cell lung cancer. Our datasets are available through our web interface (https://lungcellatlas.org). To illustrate its general utility, we use our cell atlas to generate predictions about cell-cell signaling and transcription factor hierarchies which we rigorously test using organoid models.


Asunto(s)
Feto , Pulmón , Humanos , Diferenciación Celular , Perfilación de la Expresión Génica , Pulmón/citología , Organogénesis , Organoides , Atlas como Asunto , Feto/citología
16.
Cell ; 185(2): 266-282.e15, 2022 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-35026153

RESUMEN

HIV-1-infected cells that persist despite antiretroviral therapy (ART) are frequently considered "transcriptionally silent," but active viral gene expression may occur in some cells, challenging the concept of viral latency. Applying an assay for profiling the transcriptional activity and the chromosomal locations of individual proviruses, we describe a global genomic and epigenetic map of transcriptionally active and silent proviral species and evaluate their longitudinal evolution in persons receiving suppressive ART. Using genome-wide epigenetic reference data, we show that proviral transcriptional activity is associated with activating epigenetic chromatin features in linear proximity of integration sites and in their inter- and intrachromosomal contact regions. Transcriptionally active proviruses were actively selected against during prolonged ART; however, this pattern was violated by large clones of virally infected cells that may outcompete negative selection forces through elevated intrinsic proliferative activity. Our results suggest that transcriptionally active proviruses are dynamically evolving under selection pressure by host factors.


Asunto(s)
VIH-1/genética , Provirus/genética , Transcripción Genética , Anciano , Secuencia de Bases , Evolución Biológica , Cromatina/metabolismo , Células Clonales , ADN Viral/genética , Epigénesis Genética/efectos de los fármacos , Femenino , Humanos , Ionomicina/farmacología , Masculino , Persona de Mediana Edad , Filogenia , Provirus/efectos de los fármacos , ARN Viral/genética , Acetato de Tetradecanoilforbol/farmacología , Transcripción Genética/efectos de los fármacos , Integración Viral/genética , Latencia del Virus/efectos de los fármacos , Latencia del Virus/genética
17.
Cell ; 185(5): 916-938.e58, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-35216673

RESUMEN

Treatment of severe COVID-19 is currently limited by clinical heterogeneity and incomplete description of specific immune biomarkers. We present here a comprehensive multi-omic blood atlas for patients with varying COVID-19 severity in an integrated comparison with influenza and sepsis patients versus healthy volunteers. We identify immune signatures and correlates of host response. Hallmarks of disease severity involved cells, their inflammatory mediators and networks, including progenitor cells and specific myeloid and lymphocyte subsets, features of the immune repertoire, acute phase response, metabolism, and coagulation. Persisting immune activation involving AP-1/p38MAPK was a specific feature of COVID-19. The plasma proteome enabled sub-phenotyping into patient clusters, predictive of severity and outcome. Systems-based integrative analyses including tensor and matrix decomposition of all modalities revealed feature groupings linked with severity and specificity compared to influenza and sepsis. Our approach and blood atlas will support future drug development, clinical trial design, and personalized medicine approaches for COVID-19.


Asunto(s)
Biomarcadores/sangre , COVID-19/patología , Proteoma/análisis , Adulto , Proteínas Sanguíneas/metabolismo , COVID-19/sangre , COVID-19/virología , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Femenino , Humanos , Gripe Humana/sangre , Gripe Humana/patología , Linfocitos/inmunología , Linfocitos/metabolismo , Aprendizaje Automático , Masculino , Persona de Mediana Edad , Proteína Quinasa 14 Activada por Mitógenos/genética , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Análisis de Componente Principal , SARS-CoV-2/aislamiento & purificación , Sepsis/sangre , Sepsis/patología , Índice de Severidad de la Enfermedad , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo
18.
Cell ; 185(17): 3169-3185.e20, 2022 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-35908548

RESUMEN

Mice deficient for all ten-eleven translocation (TET) genes exhibit early gastrulation lethality. However, separating cause and effect in such embryonic failure is challenging. To isolate cell-autonomous effects of TET loss, we used temporal single-cell atlases from embryos with partial or complete mutant contributions. Strikingly, when developing within a wild-type embryo, Tet-mutant cells retain near-complete differentiation potential, whereas embryos solely comprising mutant cells are defective in epiblast to ectoderm transition with degenerated mesoderm potential. We map de-repressions of early epiblast factors (e.g., Dppa4 and Gdf3) and failure to activate multiple signaling from nascent mesoderm (Lefty, FGF, and Notch) as likely cell-intrinsic drivers of TET loss phenotypes. We further suggest loss of enhancer demethylation as the underlying mechanism. Collectively, our work demonstrates an unbiased approach for defining intrinsic and extrinsic embryonic gene function based on temporal differentiation atlases and disentangles the intracellular effects of the demethylation machinery from its broader tissue-level ramifications.


Asunto(s)
Gastrulación , Mesodermo , Animales , Diferenciación Celular/genética , Embrión de Mamíferos/metabolismo , Gastrulación/genética , Regulación del Desarrollo de la Expresión Génica , Ratones , Proteínas Nucleares/metabolismo , Transducción de Señal
19.
Annu Rev Biochem ; 90: 287-320, 2021 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-34153213

RESUMEN

The field of epigenetics has exploded over the last two decades, revealing an astonishing level of complexity in the way genetic information is stored and accessed in eukaryotes. This expansion of knowledge, which is very much ongoing, has been made possible by the availability of evermore sensitive and precise molecular tools. This review focuses on the increasingly important role that chemistry plays in this burgeoning field. In an effort to make these contributions more accessible to the nonspecialist, we group available chemical approaches into those that allow the covalent structure of the protein and DNA components of chromatin to be manipulated, those that allow the activity of myriad factors that act on chromatin to be controlled, and those that allow the covalent structure and folding of chromatin to be characterized. The application of these tools is illustrated through a series of case studies that highlight how the molecular precision afforded by chemistry is being used to establish causal biochemical relationships at the heart of epigenetic regulation.


Asunto(s)
Bioquímica/métodos , Técnicas de Química Analítica/métodos , Epigenómica/métodos , Epigenoma , Transferencia Resonante de Energía de Fluorescencia , Heterocromatina/genética , Histonas/metabolismo , Técnicas de Sonda Molecular , Biosíntesis de Proteínas , Factores de Transcripción/genética , Ubiquitinación
20.
Annu Rev Biochem ; 90: 221-244, 2021 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-33784178

RESUMEN

In 1961, Jacob and Monod proposed the operon model of gene regulation. At the model's core was the modular assembly of regulators, operators, and structural genes. To illustrate the composability of these elements, Jacob and Monod linked phenotypic diversity to the architectures of regulatory circuits. In this review, we examine how the circuit blueprints imagined by Jacob and Monod laid the foundation for the first synthetic gene networks that launched the field of synthetic biology in 2000. We discuss the influences of the operon model and its broader theoretical framework on the first generation of synthetic biological circuits, which were predominantly transcriptional and posttranscriptional circuits. We also describe how recent advances in molecular biology beyond the operon model-namely, programmable DNA- and RNA-binding molecules as well as models of epigenetic and posttranslational regulation-are expanding the synthetic biology toolkit and enabling the design of more complex biological circuits.


Asunto(s)
Epigenómica/métodos , Operón , Proteínas/genética , Biología Sintética/métodos , Sistemas CRISPR-Cas , Retroalimentación Fisiológica , Regulación de la Expresión Génica , Biología Molecular/métodos , Proteínas/metabolismo , ARN Mensajero/genética , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA