Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.725
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 186(18): 3776-3792.e16, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37478861

RESUMEN

In vitro stem cell models that replicate human gastrulation have been generated, but they lack the essential extraembryonic cells needed for embryonic development, morphogenesis, and patterning. Here, we describe a robust and efficient method that prompts human extended pluripotent stem cells to self-organize into embryo-like structures, termed peri-gastruloids, which encompass both embryonic (epiblast) and extraembryonic (hypoblast) tissues. Although peri-gastruloids are not viable due to the exclusion of trophoblasts, they recapitulate critical stages of human peri-gastrulation development, such as forming amniotic and yolk sac cavities, developing bilaminar and trilaminar embryonic discs, specifying primordial germ cells, initiating gastrulation, and undergoing early neurulation and organogenesis. Single-cell RNA-sequencing unveiled transcriptomic similarities between advanced human peri-gastruloids and primary peri-gastrulation cell types found in humans and non-human primates. This peri-gastruloid platform allows for further exploration beyond gastrulation and may potentially aid in the development of human fetal tissues for use in regenerative medicine.


Asunto(s)
Implantación del Embrión , Gastrulación , Células Madre Pluripotentes , Animales , Femenino , Humanos , Embarazo , Diferenciación Celular , Embrión de Mamíferos , Desarrollo Embrionario , Organogénesis , Células Madre Pluripotentes/metabolismo , Primates
2.
Cell ; 186(15): 3166-3181.e18, 2023 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-37413989

RESUMEN

Proper preimplantation development is essential to assemble a blastocyst capable of implantation. Live imaging has uncovered major events driving early development in mouse embryos; yet, studies in humans have been limited by restrictions on genetic manipulation and lack of imaging approaches. We have overcome this barrier by combining fluorescent dyes with live imaging to reveal the dynamics of chromosome segregation, compaction, polarization, blastocyst formation, and hatching in the human embryo. We also show that blastocyst expansion mechanically constrains trophectoderm cells, causing nuclear budding and DNA shedding into the cytoplasm. Furthermore, cells with lower perinuclear keratin levels are more prone to undergo DNA loss. Moreover, applying trophectoderm biopsy, a mechanical procedure performed clinically for genetic testing, increases DNA shedding. Thus, our work reveals distinct processes underlying human development compared with mouse and suggests that aneuploidies in human embryos may not only originate from chromosome segregation errors during mitosis but also from nuclear DNA shedding.


Asunto(s)
Diagnóstico Preimplantación , Embarazo , Femenino , Humanos , Animales , Ratones , Diagnóstico Preimplantación/métodos , Blastocisto , Implantación del Embrión , Pruebas Genéticas/métodos , Aneuploidia , Biopsia/métodos
3.
Cell ; 186(1): 80-97.e26, 2023 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-36608661

RESUMEN

Glucose is a universal bioenergy source; however, its role in controlling protein interactions is unappreciated, as are its actions during differentiation-associated intracellular glucose elevation. Azido-glucose click chemistry identified glucose binding to a variety of RNA binding proteins (RBPs), including the DDX21 RNA helicase, which was found to be essential for epidermal differentiation. Glucose bound the ATP-binding domain of DDX21, altering protein conformation, inhibiting helicase activity, and dissociating DDX21 dimers. Glucose elevation during differentiation was associated with DDX21 re-localization from the nucleolus to the nucleoplasm where DDX21 assembled into larger protein complexes containing RNA splicing factors. DDX21 localized to specific SCUGSDGC motif in mRNA introns in a glucose-dependent manner and promoted the splicing of key pro-differentiation genes, including GRHL3, KLF4, OVOL1, and RBPJ. These findings uncover a biochemical mechanism of action for glucose in modulating the dimerization and function of an RNA helicase essential for tissue differentiation.


Asunto(s)
ARN Helicasas DEAD-box , Glucosa , Queratinocitos , Nucléolo Celular/metabolismo , Núcleo Celular/metabolismo , ARN Helicasas DEAD-box/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Glucosa/metabolismo , Queratinocitos/citología , Queratinocitos/metabolismo , Humanos
4.
Cell ; 182(2): 497-514.e22, 2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32579974

RESUMEN

To define the cellular composition and architecture of cutaneous squamous cell carcinoma (cSCC), we combined single-cell RNA sequencing with spatial transcriptomics and multiplexed ion beam imaging from a series of human cSCCs and matched normal skin. cSCC exhibited four tumor subpopulations, three recapitulating normal epidermal states, and a tumor-specific keratinocyte (TSK) population unique to cancer, which localized to a fibrovascular niche. Integration of single-cell and spatial data mapped ligand-receptor networks to specific cell types, revealing TSK cells as a hub for intercellular communication. Multiple features of potential immunosuppression were observed, including T regulatory cell (Treg) co-localization with CD8 T cells in compartmentalized tumor stroma. Finally, single-cell characterization of human tumor xenografts and in vivo CRISPR screens identified essential roles for specific tumor subpopulation-enriched gene networks in tumorigenesis. These data define cSCC tumor and stromal cell subpopulations, the spatial niches where they interact, and the communicating gene networks that they engage in cancer.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Genómica/métodos , Neoplasias Cutáneas/metabolismo , Animales , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Humanos , Queratinocitos/citología , Queratinocitos/metabolismo , Ratones , RNA-Seq , Análisis de la Célula Individual , Piel/metabolismo , Neoplasias Cutáneas/patología , Transcriptoma , Trasplante Heterólogo
5.
Cell ; 169(3): 407-421.e16, 2017 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-28431242

RESUMEN

The phosphorylation of agonist-occupied G-protein-coupled receptors (GPCRs) by GPCR kinases (GRKs) functions to turn off G-protein signaling and turn on arrestin-mediated signaling. While a structural understanding of GPCR/G-protein and GPCR/arrestin complexes has emerged in recent years, the molecular architecture of a GPCR/GRK complex remains poorly defined. We used a comprehensive integrated approach of cross-linking, hydrogen-deuterium exchange mass spectrometry (MS), electron microscopy, mutagenesis, molecular dynamics simulations, and computational docking to analyze GRK5 interaction with the ß2-adrenergic receptor (ß2AR). These studies revealed a dynamic mechanism of complex formation that involves large conformational changes in the GRK5 RH/catalytic domain interface upon receptor binding. These changes facilitate contacts between intracellular loops 2 and 3 and the C terminus of the ß2AR with the GRK5 RH bundle subdomain, membrane-binding surface, and kinase catalytic cleft, respectively. These studies significantly contribute to our understanding of the mechanism by which GRKs regulate the function of activated GPCRs. PAPERCLIP.


Asunto(s)
Quinasa 5 del Receptor Acoplado a Proteína-G/química , Mamíferos/metabolismo , Receptores Adrenérgicos beta 2/química , Animales , Camélidos del Nuevo Mundo , Bovinos , Quinasa 5 del Receptor Acoplado a Proteína-G/genética , Quinasa 5 del Receptor Acoplado a Proteína-G/metabolismo , Humanos , Espectrometría de Masas , Microscopía Electrónica , Modelos Moleculares , Simulación de Dinámica Molecular , Unión Proteica , Ratas , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo
6.
Cell ; 168(3): 377-389.e12, 2017 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-28129538

RESUMEN

The prototypical hallucinogen LSD acts via serotonin receptors, and here we describe the crystal structure of LSD in complex with the human serotonin receptor 5-HT2B. The complex reveals conformational rearrangements to accommodate LSD, providing a structural explanation for the conformational selectivity of LSD's key diethylamide moiety. LSD dissociates exceptionally slow from both 5-HT2BR and 5-HT2AR-a major target for its psychoactivity. Molecular dynamics (MD) simulations suggest that LSD's slow binding kinetics may be due to a "lid" formed by extracellular loop 2 (EL2) at the entrance to the binding pocket. A mutation predicted to increase the mobility of this lid greatly accelerates LSD's binding kinetics and selectively dampens LSD-mediated ß-arrestin2 recruitment. This study thus reveals an unexpected binding mode of LSD; illuminates key features of its kinetics, stereochemistry, and signaling; and provides a molecular explanation for LSD's actions at human serotonin receptors. PAPERCLIP.


Asunto(s)
Dietilamida del Ácido Lisérgico/química , Receptor de Serotonina 5-HT2B/química , Arrestina/química , Cristalografía por Rayos X , Humanos , Cinética , Modelos Químicos , Simulación de Dinámica Molecular
7.
Cell ; 170(3): 564-576.e16, 2017 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-28753430

RESUMEN

Most human epithelial tumors harbor numerous alterations, making it difficult to predict which genes are required for tumor survival. To systematically identify cancer dependencies, we analyzed 501 genome-scale loss-of-function screens performed in diverse human cancer cell lines. We developed DEMETER, an analytical framework that segregates on- from off-target effects of RNAi. 769 genes were differentially required in subsets of these cell lines at a threshold of six SDs from the mean. We found predictive models for 426 dependencies (55%) by nonlinear regression modeling considering 66,646 molecular features. Many dependencies fall into a limited number of classes, and unexpectedly, in 82% of models, the top biomarkers were expression based. We demonstrated the basis behind one such predictive model linking hypermethylation of the UBB ubiquitin gene to a dependency on UBC. Together, these observations provide a foundation for a cancer dependency map that facilitates the prioritization of therapeutic targets.


Asunto(s)
Neoplasias/genética , Neoplasias/patología , Línea Celular Tumoral , Humanos , Interferencia de ARN , Programas Informáticos , Ubiquitina/genética
8.
Cell ; 164(4): 644-55, 2016 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-26871630

RESUMEN

Repair of DNA double-strand breaks (DSBs) by non-homologous end joining is critical for neural development, and brain cells frequently contain somatic genomic variations that might involve DSB intermediates. We now use an unbiased, high-throughput approach to identify genomic regions harboring recurrent DSBs in primary neural stem/progenitor cells (NSPCs). We identify 27 recurrent DSB clusters (RDCs), and remarkably, all occur within gene bodies. Most of these NSPC RDCs were detected only upon mild, aphidicolin-induced replication stress, providing a nucleotide-resolution view of replication-associated genomic fragile sites. The vast majority of RDCs occur in long, transcribed, and late-replicating genes. Moreover, almost 90% of identified RDC-containing genes are involved in synapse function and/or neural cell adhesion, with a substantial fraction also implicated in tumor suppression and/or mental disorders. Our characterization of NSPC RDCs reveals a basis of gene fragility and suggests potential impacts of DNA breaks on neurodevelopment and neural functions.


Asunto(s)
Roturas del ADN , Células-Madre Neurales/metabolismo , Animales , Afidicolina/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Encéfalo/citología , Adhesión Celular , Moléculas de Adhesión Celular Neuronal/metabolismo , Roturas del ADN/efectos de los fármacos , Reparación del ADN por Unión de Extremidades , Reparación del ADN , Proteínas Ligadas a GPI/metabolismo , Genoma , Humanos , Ratones , Proteínas del Tejido Nervioso/metabolismo , Sinapsis , Factores de Transcripción/metabolismo , Translocación Genética
9.
Cell ; 163(4): 947-59, 2015 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-26593423

RESUMEN

RAG initiates antibody V(D)J recombination in developing lymphocytes by generating "on-target" DNA breaks at matched pairs of bona fide recombination signal sequences (RSSs). We employ bait RAG-generated breaks in endogenous or ectopically inserted RSS pairs to identify huge numbers of RAG "off-target" breaks. Such breaks occur at the simple CAC motif that defines the RSS cleavage site and are largely confined within convergent CTCF-binding element (CBE)-flanked loop domains containing bait RSS pairs. Marked orientation dependence of RAG off-target activity within loops spanning up to 2 megabases implies involvement of linear tracking. In this regard, major RAG off-targets in chromosomal translocations occur as convergent RSS pairs at enhancers within a loop. Finally, deletion of a CBE-based IgH locus element disrupts V(D)J recombination domains and, correspondingly, alters RAG on- and off-target distributions within IgH. Our findings reveal how RAG activity is developmentally focused and implicate mechanisms by which chromatin domains harness biological processes within them.


Asunto(s)
Cromosomas de los Mamíferos/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Recombinación V(D)J , Animales , Factor de Unión a CCCTC , Cromosomas de los Mamíferos/química , Proteínas de Unión al ADN/metabolismo , Genes myc , Genoma , Secuenciación de Nucleótidos de Alto Rendimiento , Proteínas de Homeodominio/metabolismo , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Linfoma/genética , Ratones , Motivos de Nucleótidos , Proteínas Represoras/metabolismo , Análisis de Secuencia de ADN , Translocación Genética
10.
Cell ; 163(5): 1124-1137, 2015 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-26582132

RESUMEN

In activated B lymphocytes, AID initiates antibody variable (V) exon somatic hypermutation (SHM) for affinity maturation in germinal centers (GCs) and IgH switch (S) region DNA breaks (DSBs) for class-switch recombination (CSR). To resolve long-standing questions, we have developed an in vivo assay to study AID targeting of passenger sequences replacing a V exon. First, we find AID targets SHM hotspots within V exon and S region passengers at similar frequencies and that the normal SHM process frequently generates deletions, indicating that SHM and CSR employ the same mechanism. Second, AID mutates targets in diverse non-Ig passengers in GC B cells at levels similar to those of V exons, definitively establishing the V exon location as "privileged" for SHM. Finally, Peyer's patch GC B cells generate a reservoir of V exons that are highly mutated before selection for affinity maturation. We discuss the implications of these findings for harnessing antibody diversification mechanisms.


Asunto(s)
Linfocitos B/metabolismo , Citidina Desaminasa/genética , Cambio de Clase de Inmunoglobulina , Hipermutación Somática de Inmunoglobulina , Recombinación V(D)J , Animales , Humanos , Ratones , Mutación , Globinas beta/genética
12.
Cell ; 159(7): 1538-48, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25483776

RESUMEN

Activation-induced cytidine deaminase (AID) initiates both somatic hypermutation (SHM) for antibody affinity maturation and DNA breakage for antibody class switch recombination (CSR) via transcription-dependent cytidine deamination of single-stranded DNA targets. Though largely specific for immunoglobulin genes, AID also acts on a limited set of off-targets, generating oncogenic translocations and mutations that contribute to B cell lymphoma. How AID is recruited to off-targets has been a long-standing mystery. Based on deep GRO-seq studies of mouse and human B lineage cells activated for CSR or SHM, we report that most robust AID off-target translocations occur within highly focal regions of target genes in which sense and antisense transcription converge. Moreover, we found that such AID-targeting "convergent" transcription arises from antisense transcription that emanates from super-enhancers within sense transcribed gene bodies. Our findings provide an explanation for AID off-targeting to a small subset of mostly lineage-specific genes in activated B cells.


Asunto(s)
Citidina Desaminasa/metabolismo , Elementos de Facilitación Genéticos , Inestabilidad Genómica , Transcripción Genética , Animales , Linfocitos B/metabolismo , Humanos , Cambio de Clase de Inmunoglobulina , Ratones , Sitio de Iniciación de la Transcripción
13.
Cell ; 157(3): 595-610, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24766807

RESUMEN

PTEN dysfunction plays a crucial role in the pathogenesis of hereditary and sporadic cancers. Here, we show that PTEN homodimerizes and, in this active conformation, exerts lipid phosphatase activity on PtdIns(3,4,5)P3. We demonstrate that catalytically inactive cancer-associated PTEN mutants heterodimerize with wild-type PTEN and constrain its phosphatase activity in a dominant-negative manner. To study the consequences of homo- and heterodimerization of wild-type and mutant PTEN in vivo, we generated Pten knockin mice harboring two cancer-associated PTEN mutations (PtenC124S and PtenG129E). Heterozygous Pten(C124S/+) and Pten(G129E/+) cells and tissues exhibit increased sensitivity to PI3-K/Akt activation compared to wild-type and Pten(+/-) counterparts, whereas this difference is no longer apparent between Pten(C124S/-) and Pten(-/-) cells. Notably, Pten KI mice are more tumor prone and display features reminiscent of complete Pten loss. Our findings reveal that PTEN loss and PTEN mutations are not synonymous and define a working model for the function and regulation of PTEN.


Asunto(s)
Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Transducción de Señal , Animales , Embrión de Mamíferos/citología , Femenino , Humanos , Pérdida de Heterocigocidad , Masculino , Ratones , Mutación , Multimerización de Proteína , Proteínas Proto-Oncogénicas c-akt/metabolismo
14.
Annu Rev Genet ; 54: 511-537, 2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-32926793

RESUMEN

Tuberculosis claims more human lives than any other bacterial infectious disease and represents a clear and present danger to global health as new tools for vaccination, treatment, and interruption of transmission have been slow to emerge. Additionally, tuberculosis presents with notable clinical heterogeneity, which complicates diagnosis, treatment, and the establishment of nonrelapsing cure. How this heterogeneity is driven by the diversity ofclinical isolates of the causative agent, Mycobacterium tuberculosis, has recently garnered attention. Herein, we review advances in the understanding of how naturally occurring variation in clinical isolates affects transmissibility, pathogenesis, immune modulation, and drug resistance. We also summarize how specific changes in transcriptional responses can modulate infection or disease outcome, together with strain-specific effects on gene essentiality. Further understanding of how this diversity of M. tuberculosis isolates affects disease and treatment outcomes will enable the development of more effective therapeutic options and vaccines for this dreaded disease.


Asunto(s)
Variación Genética/genética , Mycobacterium tuberculosis/genética , Animales , Genotipo , Humanos , Transcripción Genética/genética , Tuberculosis/microbiología
15.
Mol Cell ; 80(2): 279-295.e8, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33065020

RESUMEN

The PTEN tumor suppressor controls cell death and survival by regulating functions of various molecular targets. While the role of PTEN lipid-phosphatase activity on PtdIns(3,4,5)P3 and inhibition of PI3K pathway is well characterized, the biological relevance of PTEN protein-phosphatase activity remains undefined. Here, using knockin (KI) mice harboring cancer-associated and functionally relevant missense mutations, we show that although loss of PTEN lipid-phosphatase function cooperates with oncogenic PI3K to promote rapid mammary tumorigenesis, the additional loss of PTEN protein-phosphatase activity triggered an extensive cell death response evident in early and advanced mammary tumors. Omics and drug-targeting studies revealed that PI3Ks act to reduce glucocorticoid receptor (GR) levels, which are rescued by loss of PTEN protein-phosphatase activity to restrain cell survival. Thus, we find that the dual regulation of GR by PI3K and PTEN functions as a rheostat that can be exploited for the treatment of PTEN loss-driven cancers.


Asunto(s)
Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Fosfohidrolasa PTEN/metabolismo , Receptores de Glucocorticoides/metabolismo , Animales , Carcinogénesis , Muerte Celular , Línea Celular Tumoral , Proliferación Celular , Dexametasona/farmacología , Femenino , Humanos , Isoenzimas/metabolismo , Ratones , Modelos Biológicos , Mutación/genética , Organoides/patología , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Estabilidad Proteica , Proteoma/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
16.
Cell ; 149(1): 49-62, 2012 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-22401813

RESUMEN

Decremental loss of PTEN results in cancer susceptibility and tumor progression. PTEN elevation might therefore be an attractive option for cancer prevention and therapy. We have generated several transgenic mouse lines with PTEN expression elevated to varying levels by taking advantage of bacterial artificial chromosome (BAC)-mediated transgenesis. The "Super-PTEN" mutants are viable and show reduced body size due to decreased cell number, with no effect on cell size. Unexpectedly, PTEN elevation at the organism level results in healthy metabolism characterized by increased energy expenditure and reduced body fat accumulation. Cells derived from these mice show reduced glucose and glutamine uptake and increased mitochondrial oxidative phosphorylation and are resistant to oncogenic transformation. Mechanistically we find that PTEN elevation orchestrates this metabolic switch by regulating PI3K-dependent and -independent pathways and negatively impacting two of the most pronounced metabolic features of tumor cells: glutaminolysis and the Warburg effect.


Asunto(s)
Fosfohidrolasa PTEN/metabolismo , Transducción de Señal , Animales , Tamaño Corporal , Recuento de Células , Proliferación Celular , Respiración de la Célula , Metabolismo Energético , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo
17.
Hum Mol Genet ; 2024 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-38850567

RESUMEN

Alterations in Dp71 expression, the most ubiquitous dystrophin isoform, have been associated with patient survival across tumours. Intriguingly, in certain malignancies, Dp71 acts as a tumour suppressor, while manifesting oncogenic properties in others. This diversity could be explained by the expression of two Dp71 splice variants encoding proteins with distinct C-termini, each with specific properties. Expression of these variants has impeded the exploration of their unique roles. Using CRISPR/Cas9, we ablated the Dp71f variant with the alternative C-terminus in a sarcoma cell line not expressing the canonical C-terminal variant, and conducted molecular (RNAseq) and functional characterisation of the knockout cells. Dp71f ablation induced major transcriptomic alterations, particularly affecting the expression of genes involved in calcium signalling and ECM-receptor interaction pathways. The genome-scale metabolic analysis identified significant downregulation of glucose transport via membrane vesicle reaction (GLCter) and downregulated glycolysis/gluconeogenesis pathway. Functionally, these molecular changes corresponded with, increased calcium responses, cell adhesion, proliferation, survival under serum starvation and chemotherapeutic resistance. Knockout cells showed reduced GLUT1 protein expression, survival without attachment and their migration and invasion in vitro and in vivo were unaltered, despite increased matrix metalloproteinases release. Our findings emphasise the importance of alternative splicing of dystrophin transcripts and underscore the role of the Dp71f variant, which appears to govern distinct cellular processes frequently dysregulated in tumour cells. The loss of this regulatory mechanism promotes sarcoma cell survival and treatment resistance. Thus, Dp71f is a target for future investigations exploring the intricate functions of specific DMD transcripts in physiology and across malignancies.

18.
Nat Methods ; 19(8): 959-968, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35927480

RESUMEN

DNA-protein interactions mediate physiologic gene regulation and may be altered by DNA variants linked to polygenic disease. To enhance the speed and signal-to-noise ratio (SNR) in the identification and quantification of proteins associated with specific DNA sequences in living cells, we developed proximal biotinylation by episomal recruitment (PROBER). PROBER uses high-copy episomes to amplify SNR, and proximity proteomics (BioID) to identify the transcription factors and additional gene regulators associated with short DNA sequences of interest. PROBER quantified both constitutive and inducible association of transcription factors and corresponding chromatin regulators to target DNA sequences and binding quantitative trait loci due to single-nucleotide variants. PROBER identified alterations in regulator associations due to cancer hotspot mutations in the hTERT promoter, indicating that these mutations increase promoter association with specific gene activators. PROBER provides an approach to rapidly identify proteins associated with specific DNA sequences and their variants in living cells.


Asunto(s)
Cromatina , ADN , Biotinilación , Cromatina/genética , ADN/genética , ADN/metabolismo , Plásmidos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
Cell ; 142(3): 468-79, 2010 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-20691905

RESUMEN

Hyperactivity of mTORC1, a key mediator of cell growth, leads to stem cell depletion, although the underlying mechanisms are poorly defined. Using spermatogonial progenitor cells (SPCs) as a model system, we show that mTORC1 impairs stem cell maintenance by a negative feedback from mTORC1 to receptors required to transduce niche-derived signals. We find that SPCs lacking Plzf, a transcription factor essential for SPC maintenance, have enhanced mTORC1 activity. Aberrant mTORC1 activation in Plzf(-/-) SPCs inhibits their response to GDNF, a growth factor critical for SPC self-renewal, via negative feedback at the level of the GDNF receptor. Plzf opposes mTORC1 activity by inducing expression of the mTORC1 inhibitor Redd1. Thus, we identify the mTORC1-Plzf functional interaction as a critical rheostat for maintenance of the spermatogonial pool and propose a model whereby negative feedback from mTORC1 to the GDNF receptor balances SPC growth with self-renewal.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/metabolismo , Espermatogonias/citología , Células Madre/citología , Factores de Transcripción/metabolismo , Animales , Retroalimentación Fisiológica , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Complejos Multiproteicos , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Proteínas , Transducción de Señal , Espermatogonias/metabolismo , Células Madre/metabolismo , Serina-Treonina Quinasas TOR , Testículo/citología
20.
Nature ; 629(8013): 755-756, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38769451
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA