Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 14(1): 488, 2023 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-36717582

RESUMEN

Induced pluripotent stem cell (iPSC) reprogramming is inefficient and understanding the molecular mechanisms underlying this inefficiency holds the key to successfully control cellular identity. Here, we report 24 reprogramming roadblock genes identified by CRISPR/Cas9-mediated genome-wide knockout (KO) screening. Of these, depletion of the predicted KRAB zinc finger protein (KRAB-ZFP) Zfp266 strongly and consistently enhances murine iPSC generation in several reprogramming settings, emerging as the most robust roadblock. We show that ZFP266 binds Short Interspersed Nuclear Elements (SINEs) adjacent to binding sites of pioneering factors, OCT4 (POU5F1), SOX2, and KLF4, and impedes chromatin opening. Replacing the KRAB co-suppressor with co-activator domains converts ZFP266 from an inhibitor to a potent facilitator of iPSC reprogramming. We propose that the SINE-KRAB-ZFP interaction is a critical regulator of chromatin accessibility at regulatory elements required for efficient cellular identity changes. In addition, this work serves as a resource to further illuminate molecular mechanisms hindering reprogramming.


Asunto(s)
Células Madre Pluripotentes Inducidas , Dedos de Zinc , Animales , Ratones , Reprogramación Celular/genética , Cromatina/genética , Cromatina/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Factor 4 Similar a Kruppel
2.
Genes Dev ; 31(20): 2085-2098, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-29138277

RESUMEN

Expression of the transcription factors OCT4, SOX2, KLF4, and cMYC (OSKM) reprograms somatic cells into induced pluripotent stem cells (iPSCs). Reprogramming is a slow and inefficient process, suggesting the presence of safeguarding mechanisms that counteract cell fate conversion. One such mechanism is senescence. To identify modulators of reprogramming-induced senescence, we performed a genome-wide shRNA screen in primary human fibroblasts expressing OSKM. In the screen, we identified novel mediators of OSKM-induced senescence and validated previously implicated genes such as CDKN1A We developed an innovative approach that integrates single-cell RNA sequencing (scRNA-seq) with the shRNA screen to investigate the mechanism of action of the identified candidates. Our data unveiled regulation of senescence as a novel way by which mechanistic target of rapamycin (mTOR) influences reprogramming. On one hand, mTOR inhibition blunts the induction of cyclin-dependent kinase (CDK) inhibitors (CDKIs), including p16INK4a, p21CIP1, and p15INK4b, preventing OSKM-induced senescence. On the other hand, inhibition of mTOR blunts the senescence-associated secretory phenotype (SASP), which itself favors reprogramming. These contrasting actions contribute to explain the complex effect that mTOR has on reprogramming. Overall, our study highlights the advantage of combining functional screens with scRNA-seq to accelerate the discovery of pathways controlling complex phenotypes.


Asunto(s)
Reprogramación Celular , Senescencia Celular , Perfilación de la Expresión Génica , ARN Interferente Pequeño , Análisis de Secuencia de ARN , Serina-Treonina Quinasas TOR/fisiología , Factores de Transcripción/metabolismo , Animales , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Factor 4 Similar a Kruppel , Ratones , Análisis de la Célula Individual , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
3.
Cell Stem Cell ; 21(6): 791-805.e9, 2017 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-29174331

RESUMEN

Reprogramming of cellular identity using exogenous expression of transcription factors (TFs) is a powerful and exciting tool for tissue engineering, disease modeling, and regenerative medicine. However, generation of desired cell types using this approach is often plagued by inefficiency, slow conversion, and an inability to produce mature functional cells. Here, we show that expression of constitutively active SMAD2/3 significantly improves the efficiency of induced pluripotent stem cell (iPSC) generation by the Yamanaka factors. Mechanistically, SMAD3 interacts with reprogramming factors and co-activators and co-occupies OCT4 target loci during reprogramming. Unexpectedly, active SMAD2/3 also markedly enhances three other TF-mediated direct reprogramming conversions, from B cells to macrophages, myoblasts to adipocytes, and human fibroblasts to neurons, highlighting broad and general roles for SMAD2/3 as cell-reprogramming potentiators. Our results suggest that co-expression of active SMAD2/3 could enhance multiple types of TF-based cell identity conversion and therefore be a powerful tool for cellular engineering.


Asunto(s)
Reprogramación Celular , Células Madre Pluripotentes Inducidas/metabolismo , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factores de Transcripción/metabolismo , Línea Celular , Humanos , Factores de Transcripción/genética
4.
Science ; 351(6274): aad5510, 2016 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-26797145

RESUMEN

Differentiated macrophages can self-renew in tissues and expand long term in culture, but the gene regulatory mechanisms that accomplish self-renewal in the differentiated state have remained unknown. Here we show that in mice, the transcription factors MafB and c-Maf repress a macrophage-specific enhancer repertoire associated with a gene network that controls self-renewal. Single-cell analysis revealed that, in vivo, proliferating resident macrophages can access this network by transient down-regulation of Maf transcription factors. The network also controls embryonic stem cell self-renewal but is associated with distinct embryonic stem cell-specific enhancers. This indicates that distinct lineage-specific enhancer platforms regulate a shared network of genes that control self-renewal potential in both stem and mature cells.


Asunto(s)
Diferenciación Celular/genética , Linaje de la Célula/genética , Células Madre Embrionarias/citología , Elementos de Facilitación Genéticos/fisiología , Regulación de la Expresión Génica , Macrófagos/citología , Animales , Proliferación Celular , Células Cultivadas , Regulación hacia Abajo , Redes Reguladoras de Genes , Factor de Transcripción MafB/metabolismo , Ratones , Proteínas Proto-Oncogénicas c-maf/metabolismo , Análisis de la Célula Individual , Activación Transcripcional
5.
BMC Microbiol ; 10: 190, 2010 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-20624274

RESUMEN

BACKGROUND: Streptococcus pneumoniae is a widely distributed commensal Gram-positive bacteria of the upper respiratory tract. Pneumococcal colonization can progress to invasive disease, and thus become lethal, reason why antibiotics and vaccines are designed to limit the dramatic effects of the bacteria in such cases. As a consequence, pneumococcus has developed efficient antibiotic resistance, and the use of vaccines covering a limited number of serotypes such as Pneumovax and Prevnar results in the expansion of non-covered serotypes. Pneumococcal surface proteins represent challenging candidates for the development of new therapeutic targets against the bacteria. Despite the number of described virulence factors, we believe that the majority of them remain to be characterized. This is the reason why pneumococcus invasion processes are still largely unknown. RESULTS: Availability of genome sequences facilitated the identification of pneumococcal surface proteins bearing characteristic motifs such as choline-binding proteins (Cbp) and peptidoglycan binding (LPXTG) proteins. We designed a medium throughput approach to systematically test for interactions between these pneumococcal surface proteins and host proteins (extracellular matrix proteins, circulating proteins or immunity related proteins). We cloned, expressed and purified 28 pneumococcal surface proteins. Interactions were tested in a solid phase assay, which led to the identification of 23 protein-protein interactions among which 20 are new. CONCLUSIONS: We conclude that whether peptidoglycan binding proteins do not appear to be major adhesins, most of the choline-binding proteins interact with host proteins (elastin and C reactive proteins are the major Cbp partners). These newly identified interactions open the way to a better understanding of host-pneumococcal interactions.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas de la Membrana/metabolismo , Infecciones Neumocócicas/metabolismo , Streptococcus pneumoniae/metabolismo , Adhesinas Bacterianas/genética , Proteínas Bacterianas/genética , Humanos , Proteínas de la Membrana/genética , Infecciones Neumocócicas/microbiología , Unión Proteica , Streptococcus pneumoniae/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...