Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Nature ; 616(7956): 365-372, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37020022

RESUMEN

Human gene expression is regulated by more than 2,000 transcription factors and chromatin regulators1,2. Effector domains within these proteins can activate or repress transcription. However, for many of these regulators we do not know what type of effector domains they contain, their location in the protein, their activation and repression strengths, and the sequences that are necessary for their functions. Here, we systematically measure the effector activity of more than 100,000 protein fragments tiling across most chromatin regulators and transcription factors in human cells (2,047 proteins). By testing the effect they have when recruited at reporter genes, we annotate 374 activation domains and 715 repression domains, roughly 80% of which are new and have not been previously annotated3-5. Rational mutagenesis and deletion scans across all the effector domains reveal aromatic and/or leucine residues interspersed with acidic, proline, serine and/or glutamine residues are necessary for activation domain activity. Furthermore, most repression domain sequences contain sites for small ubiquitin-like modifier (SUMO)ylation, short interaction motifs for recruiting corepressors or are structured binding domains for recruiting other repressive proteins. We discover bifunctional domains that can both activate and repress, some of which dynamically split a cell population into high- and low-expression subpopulations. Our systematic annotation and characterization of effector domains provide a rich resource for understanding the function of human transcription factors and chromatin regulators, engineering compact tools for controlling gene expression and refining predictive models of effector domain function.


Asunto(s)
Regulación de la Expresión Génica , Mutagénesis , Dominios Proteicos , Factores de Transcripción , Transcripción Genética , Humanos , Cromatina/genética , Cromatina/metabolismo , Genes Reporteros/genética , Factores de Transcripción/química , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Dominios Proteicos/genética , Proteínas Represoras/química , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Sumoilación
2.
Development ; 146(12)2019 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-31249006

RESUMEN

Understanding chromatin regulation holds enormous promise for controlling gene regulation, predicting cellular identity, and developing diagnostics and cellular therapies. However, the dynamic nature of chromatin, together with cell-to-cell heterogeneity in its structure, limits our ability to extract its governing principles. Single cell mapping of chromatin modifications, in conjunction with expression measurements, could help overcome these limitations. Here, we review recent advances in single cell-based measurements of chromatin modifications, including optimization to reduce DNA loss, improved DNA sequencing, barcoding, and antibody engineering. We also highlight several applications of these techniques that have provided insights into cell-type classification, mapping modification co-occurrence and heterogeneity, and monitoring chromatin dynamics.


Asunto(s)
Cromatina/química , Análisis de la Célula Individual/métodos , Acetilación , Animales , Anticuerpos/química , Islas de CpG , ADN/química , Metilación de ADN , Reparación del ADN , Endonucleasas/metabolismo , Epigénesis Genética , Epigenoma , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ingeniería Genética/métodos , Histonas/química , Humanos , Ratones , Análisis de Secuencia de ADN
3.
Proc Natl Acad Sci U S A ; 115(40): 10172-10177, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30232263

RESUMEN

Alzheimer's disease (AD), the most common form of dementia, is characterized by the abnormal accumulation of amyloid plaques and hyperphosphorylated tau aggregates, as well as microgliosis. Hemizygous missense variants in Triggering Receptor Expressed on Myeloid Cells 2 (TREM2) are associated with elevated risk for developing late-onset AD. These variants are hypothesized to result in loss of function, mimicking TREM2 haploinsufficiency. However, the consequences of TREM2 haploinsufficiency on tau pathology and microglial function remain unknown. We report the effects of partial and complete loss of TREM2 on microglial function and tau-associated deficits. In vivo imaging revealed that microglia from aged TREM2-haploinsufficient mice show a greater impairment in their injury response compared with microglia from aged TREM2-KO mice. In transgenic mice expressing mutant human tau, TREM2 haploinsufficiency, but not complete loss of TREM2, increased tau pathology. In addition, whereas complete TREM2 deficiency protected against tau-mediated microglial activation and atrophy, TREM2 haploinsufficiency elevated expression of proinflammatory markers and exacerbated atrophy at a late stage of disease. The differential effects of partial and complete loss of TREM2 on microglial function and tau pathology provide important insights into the critical role of TREM2 in AD pathogenesis.


Asunto(s)
Enfermedad de Alzheimer , Haploinsuficiencia , Hemicigoto , Glicoproteínas de Membrana , Microglía/metabolismo , Mutación Missense , Receptores Inmunológicos , Envejecimiento/genética , Envejecimiento/metabolismo , Envejecimiento/patología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Animales , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Noqueados , Microglía/patología , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo
4.
Cell Syst ; 14(6): 482-500.e8, 2023 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-37348463

RESUMEN

Viruses encode transcriptional regulatory proteins critical for controlling viral and host gene expression. Given their multifunctional nature and high sequence divergence, it is unclear which viral proteins can affect transcription and which specific sequences contribute to this function. Using a high-throughput assay, we measured the transcriptional regulatory potential of over 60,000 protein tiles across ∼1,500 proteins from 11 coronaviruses and all nine human herpesviruses. We discovered hundreds of transcriptional effector domains, including a conserved repression domain in all coronavirus Spike homologs, dual activation-repression domains in viral interferon regulatory factors (VIRFs), and an activation domain in six herpesvirus homologs of the single-stranded DNA-binding protein that we show is important for viral replication and late gene expression in Kaposi's sarcoma-associated herpesvirus (KSHV). For the effector domains we identified, we investigated their mechanisms via high-throughput sequence and chemical perturbations, pinpointing sequence motifs essential for function. This work massively expands viral protein annotations, serving as a springboard for studying their biological and health implications and providing new candidates for compact gene regulation tools.


Asunto(s)
Herpesvirus Humano 8 , Humanos , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Replicación Viral/genética , Regulación de la Expresión Génica
5.
bioRxiv ; 2023 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-37873347

RESUMEN

Histone H3.3 is frequently mutated in cancers, with the lysine 36 to methionine mutation (K36M) being a hallmark of chondroblastomas. While it is known that H3.3K36M changes the cellular epigenetic landscape, it remains unclear how it affects the dynamics of gene expression. Here, we use a synthetic reporter to measure the effect of H3.3K36M on silencing and epigenetic memory after recruitment of KRAB: a member of the largest class of human repressors, commonly used in synthetic biology, and associated with H3K9me3. We find that H3.3K36M, which decreases H3K36 methylation, leads to a decrease in epigenetic memory and promoter methylation weeks after KRAB release. We propose a new model for establishment and maintenance of epigenetic memory, where H3K36 methylation is necessary to convert H3K9me3 domains into DNA methylation for stable epigenetic memory. Our quantitative model can inform oncogenic mechanisms and guide development of epigenetic editing tools.

6.
Cell Syst ; 14(9): 746-763.e5, 2023 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-37543039

RESUMEN

Despite growing knowledge of the functions of individual human transcriptional effector domains, much less is understood about how multiple effector domains within the same protein combine to regulate gene expression. Here, we measure transcriptional activity for 8,400 effector domain combinations by recruiting them to reporter genes in human cells. In our assay, weak and moderate activation domains synergize to drive strong gene expression, whereas combining strong activators often results in weaker activation. In contrast, repressors combine linearly and produce full gene silencing, and repressor domains often overpower activation domains. We use this information to build a synthetic transcription factor whose function can be tuned between repression and activation independent of recruitment to target genes by using a small-molecule drug. Altogether, we outline the basic principles of how effector domains combine to regulate gene expression and demonstrate their value in building precise and flexible synthetic biology tools. A record of this paper's transparent peer review process is included in the supplemental information.


Asunto(s)
Proteínas Represoras , Transcripción Genética , Humanos , Transcripción Genética/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Regulación de la Expresión Génica/genética , Genes Reporteros
7.
Elife ; 112022 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-35678392

RESUMEN

In mammalian cells genes that are in close proximity can be transcriptionally coupled: silencing or activating one gene can affect its neighbors. Understanding these dynamics is important for natural processes, such as heterochromatin spreading during development and aging, and when designing synthetic gene regulation circuits. Here, we systematically dissect this process in single cells by recruiting and releasing repressive chromatin regulators at dual-gene synthetic reporters, and measuring how fast gene silencing and reactivation spread as a function of intergenic distance and configuration of insulator elements. We find that silencing by KRAB, associated with histone methylation, spreads between two genes within hours, with a time delay that increases with distance. This fast KRAB-mediated spreading is not blocked by the classical cHS4 insulators. Silencing by histone deacetylase HDAC4 of the upstream gene can also facilitate background silencing of the downstream gene by PRC2, but with a days-long delay that does not change with distance. This slower silencing can sometimes be stopped by insulators. Gene reactivation of neighboring genes is also coupled, with strong promoters and insulators determining the order of reactivation. Our data can be described by a model of multi-gene regulation that builds upon previous knowledge of heterochromatin spreading, where both gene silencing and gene reactivation can act at a distance, allowing for coordinated dynamics via chromatin regulator recruitment.


Asunto(s)
Cromatina , Heterocromatina , Animales , Cromatina/genética , Regulación de la Expresión Génica , Silenciador del Gen , Heterocromatina/genética , Elementos Aisladores , Mamíferos/genética
8.
Neuron ; 104(2): 239-255.e12, 2019 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-31422865

RESUMEN

CRISPR/Cas9-based functional genomics have transformed our ability to elucidate mammalian cell biology. However, most previous CRISPR-based screens were conducted in cancer cell lines rather than healthy, differentiated cells. Here, we describe a CRISPR interference (CRISPRi)-based platform for genetic screens in human neurons derived from induced pluripotent stem cells (iPSCs). We demonstrate robust and durable knockdown of endogenous genes in such neurons and present results from three complementary genetic screens. First, a survival-based screen revealed neuron-specific essential genes and genes that improved neuronal survival upon knockdown. Second, a screen with a single-cell transcriptomic readout uncovered several examples of genes whose knockdown had strikingly cell-type-specific consequences. Third, a longitudinal imaging screen detected distinct consequences of gene knockdown on neuronal morphology. Our results highlight the power of unbiased genetic screens in iPSC-derived differentiated cell types and provide a platform for systematic interrogation of normal and disease states of neurons. VIDEO ABSTRACT.


Asunto(s)
Sistemas CRISPR-Cas , Técnicas de Silenciamiento del Gen/métodos , Células Madre Pluripotentes Inducidas , Neuronas/metabolismo , Supervivencia Celular , Humanos , Microscopía Confocal , Neuronas/citología , RNA-Seq , Análisis de la Célula Individual
9.
Science ; 363(6428)2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30765536

RESUMEN

How hexanucleotide GGGGCC (G4C2) repeat expansions in C9orf72 cause frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) is not understood. We developed a mouse model engineered to express poly(PR), a proline-arginine (PR) dipeptide repeat protein synthesized from expanded G4C2 repeats. The expression of green fluorescent protein-conjugated (PR)50 (a 50-repeat PR protein) throughout the mouse brain yielded progressive brain atrophy, neuron loss, loss of poly(PR)-positive cells, and gliosis, culminating in motor and memory impairments. We found that poly(PR) bound DNA, localized to heterochromatin, and caused heterochromatin protein 1α (HP1α) liquid-phase disruptions, decreases in HP1α expression, abnormal histone methylation, and nuclear lamina invaginations. These aberrations of histone methylation, lamins, and HP1α, which regulate heterochromatin structure and gene expression, were accompanied by repetitive element expression and double-stranded RNA accumulation. Thus, we uncovered mechanisms by which poly(PR) may contribute to the pathogenesis of C9orf72-associated FTD and ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Proteína C9orf72/metabolismo , Dipéptidos/metabolismo , Heterocromatina/patología , ARN Bicatenario/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Encéfalo/metabolismo , Proteína C9orf72/genética , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/metabolismo , Dipéptidos/genética , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Lámina Nuclear/patología , Secuencias Repetitivas de Ácidos Nucleicos
10.
Stem Cell Reports ; 9(4): 1221-1233, 2017 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-28966121

RESUMEN

Lowering total tau levels is an attractive therapeutic strategy for Alzheimer's disease and other tauopathies. High-throughput screening in neurons derived from human induced pluripotent stem cells (iPSCs) is a powerful tool to identify tau-targeted therapeutics. However, such screens have been hampered by heterogeneous neuronal production, high cost and low yield, and multi-step differentiation procedures. We engineered an isogenic iPSC line that harbors an inducible neurogenin 2 transgene, a transcription factor that rapidly converts iPSCs to neurons, integrated at the AAVS1 locus. Using a simplified two-step protocol, we differentiated these iPSCs into cortical glutamatergic neurons with minimal well-to-well variability. We developed a robust high-content screening assay to identify tau-lowering compounds in LOPAC and identified adrenergic receptors agonists as a class of compounds that reduce endogenous human tau. These techniques enable the use of human neurons for high-throughput screening of drugs to treat neurodegenerative disease.


Asunto(s)
Diferenciación Celular , Descubrimiento de Drogas , Regulación de la Expresión Génica/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento , Células Madre Pluripotentes Inducidas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Proteínas tau/genética , Línea Celular , Supervivencia Celular , Células Cultivadas , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos , Expresión Génica , Orden Génico , Vectores Genéticos/genética , Glutamina/metabolismo , Humanos , Potenciales de la Membrana , Neuronas/citología , Proteínas tau/metabolismo
11.
Stem Cell Reports ; 9(6): 1745-1753, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29198827

RESUMEN

Human astrocytes network with neurons in dynamic ways that are still poorly defined. Our ability to model this relationship is hampered by the lack of relevant and convenient tools to recapitulate this complex interaction. To address this barrier, we have devised efficient coculture systems utilizing 3D organoid-like spheres, termed asteroids, containing pre-differentiated human pluripotent stem cell (hPSC)-derived astrocytes (hAstros) combined with neurons generated from hPSC-derived neural stem cells (hNeurons) or directly induced via Neurogenin 2 overexpression (iNeurons). Our systematic methods rapidly produce structurally complex hAstros and synapses in high-density coculture with iNeurons in precise numbers, allowing for improved studies of neural circuit function, disease modeling, and drug screening. We conclude that these bioengineered neural circuit model systems are reliable and scalable tools to accurately study aspects of human astrocyte-neuron functional properties while being easily accessible for cell-type-specific manipulations and observations.


Asunto(s)
Astrocitos/citología , Diferenciación Celular/genética , Técnicas de Cocultivo , Neuronas/citología , Astrocitos/metabolismo , Linaje de la Célula/genética , Linaje de la Célula/fisiología , Células Cultivadas , Humanos , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Sinapsis/metabolismo , Sinapsis/fisiología
12.
Sci Transl Med ; 9(385)2017 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-28404863

RESUMEN

Heterozygous mutations in the GRN gene lead to progranulin (PGRN) haploinsufficiency and cause frontotemporal dementia (FTD), a neurodegenerative syndrome of older adults. Homozygous GRN mutations, on the other hand, lead to complete PGRN loss and cause neuronal ceroid lipofuscinosis (NCL), a lysosomal storage disease usually seen in children. Given that the predominant clinical and pathological features of FTD and NCL are distinct, it is controversial whether the disease mechanisms associated with complete and partial PGRN loss are similar or distinct. We show that PGRN haploinsufficiency leads to NCL-like features in humans, some occurring before dementia onset. Noninvasive retinal imaging revealed preclinical retinal lipofuscinosis in heterozygous GRN mutation carriers. Increased lipofuscinosis and intracellular NCL-like storage material also occurred in postmortem cortex of heterozygous GRN mutation carriers. Lymphoblasts from heterozygous GRN mutation carriers accumulated prominent NCL-like storage material, which could be rescued by normalizing PGRN expression. Fibroblasts from heterozygous GRN mutation carriers showed impaired lysosomal protease activity. Our findings indicate that progranulin haploinsufficiency caused accumulation of NCL-like storage material and early retinal abnormalities in humans and implicate lysosomal dysfunction as a central disease process in GRN-associated FTD and GRN-associated NCL.


Asunto(s)
Haploinsuficiencia/fisiología , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Lipofuscinosis Ceroideas Neuronales/metabolismo , Lipofuscinosis Ceroideas Neuronales/patología , Animales , Células Cultivadas , Lóbulo Frontal/metabolismo , Lóbulo Frontal/ultraestructura , Demencia Frontotemporal/genética , Demencia Frontotemporal/metabolismo , Demencia Frontotemporal/patología , Haploinsuficiencia/genética , Heterocigoto , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Lisosomas , Ratones , Microscopía Electrónica , Mutación/genética , Lipofuscinosis Ceroideas Neuronales/genética , Progranulinas , Retina/metabolismo , Retina/ultraestructura
13.
J Clin Invest ; 124(3): 1027-36, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24509084

RESUMEN

Regulatory T cells (Tregs), which are characterized by expression of the transcription factor Foxp3, are a dynamic and heterogeneous population of cells that control immune responses and prevent autoimmunity. We recently identified a subset of Tregs in murine skin with properties typical of memory cells and defined this population as memory Tregs (mTregs). Due to the importance of these cells in regulating tissue inflammation in mice, we analyzed this cell population in humans and found that almost all Tregs in normal skin had an activated memory phenotype. Compared with mTregs in peripheral blood, cutaneous mTregs had unique cell surface marker expression and cytokine production. In normal human skin, mTregs preferentially localized to hair follicles and were more abundant in skin with high hair density. Sequence comparison of TCRs from conventional memory T helper cells and mTregs isolated from skin revealed little homology between the two cell populations, suggesting that they recognize different antigens. Under steady-state conditions, mTregs were nonmigratory and relatively unresponsive; however, in inflamed skin from psoriasis patients, mTregs expanded, were highly proliferative, and produced low levels of IL-17. Taken together, these results identify a subset of Tregs that stably resides in human skin and suggest that these cells are qualitatively defective in inflammatory skin disease.


Asunto(s)
Folículo Piloso/patología , Linfocitos T Reguladores/metabolismo , Adulto , Anciano , Animales , Antígenos CD/metabolismo , Movimiento Celular , Proliferación Celular , Células Cultivadas , Femenino , Factores de Transcripción Forkhead/metabolismo , Folículo Piloso/inmunología , Humanos , Memoria Inmunológica , Interleucina-17/metabolismo , Masculino , Ratones , Ratones Endogámicos NOD , Persona de Mediana Edad , Fenotipo , Psoriasis/inmunología , Psoriasis/patología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores CCR7/metabolismo , Piel/inmunología , Linfocitos T Reguladores/inmunología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA