Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
BMC Cancer ; 22(1): 164, 2022 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-35148692

RESUMEN

BACKGROUND: Exosomes play a key role in cell-to-cell communication and are integral component of the tumor microenvironment. Recent observations suggest transfer of RNA through tumor-derived exosomes that can potentially translate into regulatory proteins in the recipient cells. Role of cervical cancer-derived exosomes and their transcript cargo is poorly understood. MATERIALS AND METHODS: The total RNA of exosomes from HPV-positive (SiHa and HeLa) and HPV-negative (C33a) cervical cancer cell lines were extracted and the transcripts were estimated using Illumina HiSeq X. Further, validation of HPV transcripts were performed using RT-PCR. RESULTS: 3099 transcripts were found to be differentially-exported in HPV-positive vs. HPV-negative exosomes (p value <0.05). Analysis of top 10 GO terms and KEGG pathways showed enrichment of transcripts belonging to axon guidance and tumor innervation in HPV-positive exosomes. Among top 20 overexpressed transcripts, EVC2, LUZP1 and ANKS1B were the most notable due to their involvement in Hh signaling, cellular migration and invasion, respectively. Further, low levels of HPV-specific reads were detected. RT-PCR validation revealed presence of E6*I splice variant of HPV18 in exosomal RNA of HeLa cells. The E6*I transcripts were consistently retained in exosomes obtained from HeLa cells undergoing 5-FU and cisplatin-induced oxidative stress. CONCLUSION: Our data suggests the enrichment of poly-A RNA transcripts in the exosomal cargo of cervical cancer cells, which includes pro-tumorigenic cellular RNA and viral transcripts such as HPV E6, which may have clinical utility as potential exosomal biomarkers of cervical cancer.


Asunto(s)
Exosomas/genética , Exosomas/virología , Proteínas Oncogénicas Virales/genética , ARN Viral/genética , Neoplasias del Cuello Uterino/virología , Biomarcadores de Tumor/genética , Línea Celular Tumoral , Femenino , Perfilación de la Expresión Génica , Células HeLa , Humanos
2.
Nucleic Acids Res ; 43(3): 1513-28, 2015 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-25578969

RESUMEN

Sox2 and Pax6 are transcription factors that direct cell fate decision during neurogenesis, yet the mechanism behind how they cooperate on enhancer DNA elements and regulate gene expression is unclear. By systematically interrogating Sox2 and Pax6 interaction on minimal enhancer elements, we found that cooperative DNA recognition relies on combinatorial nucleotide switches and precisely spaced, but cryptic composite DNA motifs. Surprisingly, all tested Sox and Pax paralogs have the capacity to cooperate on such enhancer elements. NMR and molecular modeling reveal very few direct protein-protein interactions between Sox2 and Pax6, suggesting that cooperative binding is mediated by allosteric interactions propagating through DNA structure. Furthermore, we detected and validated several novel sites in the human genome targeted cooperatively by Sox2 and Pax6. Collectively, we demonstrate that Sox-Pax partnerships have the potential to substantially alter DNA target specificities and likely enable the pleiotropic and context-specific action of these cell-lineage specifiers.


Asunto(s)
ADN/fisiología , Elementos de Facilitación Genéticos , Proteínas del Ojo/fisiología , Proteínas de Homeodominio/fisiología , Factores de Transcripción Paired Box/fisiología , Proteínas Represoras/fisiología , Factores de Transcripción SOXB1/fisiología , Secuencia de Aminoácidos , Secuencia de Bases , Cartilla de ADN , Ensayo de Cambio de Movilidad Electroforética , Proteínas del Ojo/química , Proteínas de Homeodominio/química , Humanos , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/química , Proteínas Represoras/química , Factores de Transcripción SOXB1/química , Homología de Secuencia de Aminoácido
3.
Int J Biol Sci ; 17(2): 527-538, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33613110

RESUMEN

Germline specification is a fundamental step for human reproduction and this biological phenomenon possesses technical challenges to study in vivo as it occurs immediately after blastocyst implantation. The establishment of in vitro human primordial germ cell-like cells (hPGCLCs) induction system allows sophisticated characterization of human primordial germ cells (hPGCs) development. However, the underlying molecular mechanisms of hPGCLC specification are not fully elucidated. Here, we observed particularly high expression of the histone demethylase KDM2B in male fetal germ cells (FGCs) but not in male somatic cells. Besides, KDM2B shared similar expression pattern with hPGC marker genes in hPGCLCs, suggesting an important role of KDM2B in germ cell development. Although deletion of KDM2B had no significant effects on human embryonic stem cell (hESC)'s pluripotency, loss of KDM2B dramatically impaired hPGCLCs differentiation whereas ectopically expressed KDM2B could efficiently rescue such defect, indicating this defect was due to KDM2B's loss in hPGCLC specification. Mechanistically, as revealed by the transcriptional profiling, KDM2B suppressed the expression of somatic genes thus inhibited somatic differentiation during hPGCLC specification. These data collectively indicate that KDM2B is an indispensable epigenetic regulator for hPGCLC specification, shedding lights on how epigenetic regulations orchestrate transcriptional events in hPGC development for future investigation.


Asunto(s)
Diferenciación Celular/fisiología , Linaje de la Célula , Proteínas F-Box/fisiología , Células Germinativas/citología , Histona Demetilasas con Dominio de Jumonji/fisiología , Células Cultivadas , Células Madre Embrionarias/citología , Proteínas F-Box/genética , Técnicas de Silenciamiento del Gen , Humanos , Histona Demetilasas con Dominio de Jumonji/genética
4.
Stem Cell Reports ; 16(5): 1245-1261, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33930315

RESUMEN

In vitro induction of human primordial germ cell-like cells (hPGCLCs) provides an ideal platform to recapitulate hPGC development. However, the detailed molecular mechanisms regulating the induction of hPGCLCs remain largely uncharacterized. Here, we profiled the chromatin accessibility and transcriptome dynamics throughout the process of hPGCLC induction. Genetic ablation of SOX15 indicated the crucial roles of SOX15 in the maintenance of hPGCLCs. Mechanistically, SOX15 exerted its roles via suppressing somatic gene expression and sustaining latent pluripotency. Notably, ETV5, a downstream regulator of SOX15, was also uncovered to be essential for hPGCLC maintenance. Finally, a stepwise switch of OCT4/SOX2, OCT4/SOX17, and OCT4/SOX15 binding motifs were found to be enriched in closed-to-open regions of human embryonic stem cells, and early- and late-stage hPGCLCs, respectively. Collectively, our data characterized the chromatin accessibility and transcriptome landscapes throughout hPGCLC induction and defined the SOX15-mediated regulatory networks underlying this process.


Asunto(s)
Cromatina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/metabolismo , Células Madre Pluripotentes/metabolismo , Transcripción Genética , Secuencia de Bases , Linaje de la Célula/genética , Células Germinativas/citología , Células Madre Embrionarias Humanas/citología , Células Madre Embrionarias Humanas/metabolismo , Humanos , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/citología , Secuencias Reguladoras de Ácidos Nucleicos/genética , Factores de Transcripción SOX/metabolismo , Factor de Transcripción AP-2/metabolismo
5.
FEBS J ; 287(1): 122-144, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31569299

RESUMEN

The functional consequences of cancer-associated missense mutations are unclear for the majority of proteins. We have previously demonstrated that the activity of SOX and Pit-Oct-Unc (POU) family factors during pluripotency reprogramming can be switched and enhanced with rationally placed point mutations. Here, we interrogated cancer mutation databases and identified recurrently mutated positions at critical structural interfaces of the DNA-binding domains of paralogous SOX and POU family transcription factors. Using the conversion of mouse embryonic fibroblasts to induced pluripotent stem cells as functional readout, we identified several gain-of-function mutations that enhance pluripotency reprogramming by SOX2 and OCT4. Wild-type SOX17 cannot support reprogramming but the recurrent missense mutation SOX17-V118M is capable of inducing pluripotency. Furthermore, SOX17-V118M promotes oncogenic transformation, enhances thermostability and elevates cellular protein levels of SOX17. We conclude that the mutational profile of SOX and POU family factors in cancer can guide the design of high-performance reprogramming factors. Furthermore, we propose cellular reprogramming as a suitable assay to study the functional impact of cancer-associated mutations.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Pluripotentes Inducidas/citología , Mutación Missense , Neoplasias/patología , Factor 3 de Transcripción de Unión a Octámeros/genética , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXF/genética , Animales , Diferenciación Celular , Células Cultivadas , Reprogramación Celular , Células Madre Embrionarias/metabolismo , Perfilación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Neoplasias/genética , Neoplasias/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción SOXF/metabolismo
6.
Nat Commun ; 10(1): 3477, 2019 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-31375664

RESUMEN

Oct4, along with Sox2 and Klf4 (SK), can induce pluripotency but structurally similar factors like Oct6 cannot. To decode why Oct4 has this unique ability, we compare Oct4-binding, accessibility patterns and transcriptional waves with Oct6 and an Oct4 mutant defective in the dimerization with Sox2 (Oct4defSox2). We find that initial silencing of the somatic program proceeds indistinguishably with or without Oct4. Oct6 mitigates the mesenchymal-to-epithelial transition and derails reprogramming. These effects are a consequence of differences in genome-wide binding, as the early binding profile of Oct4defSox2 resembles Oct4, whilst Oct6 does not bind pluripotency enhancers. Nevertheless, in the Oct6-SK condition many otherwise Oct4-bound locations become accessible but chromatin opening is compromised when Oct4defSox2 occupies these sites. We find that Sox2 predominantly facilitates chromatin opening, whilst Oct4 serves an accessory role. Formation of Oct4/Sox2 heterodimers is essential for pluripotency establishment; however, reliance on Oct4/Sox2 heterodimers declines during pluripotency maintenance.


Asunto(s)
Reprogramación Celular/genética , Cromatina/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Factores de Transcripción SOXB1/metabolismo , Animales , Células Cultivadas , Embrión de Mamíferos , Transición Epitelial-Mesenquimal/genética , Fibroblastos , Células Madre Pluripotentes Inducidas/fisiología , Factor 4 Similar a Kruppel , Ratones Transgénicos , Mutación , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 6 de Transcripción de Unión a Octámeros/metabolismo , Cultivo Primario de Células , Multimerización de Proteína/genética , Factores de Transcripción SOXB1/genética , Factores de Tiempo
7.
Stem Cell Reports ; 11(2): 593-606, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30078555

RESUMEN

Directed biomolecular evolution is widely used to tailor and enhance enzymes, fluorescent proteins, and antibodies but has hitherto not been applied in the reprogramming of mammalian cells. Here, we describe a method termed directed evolution of reprogramming factors by cell selection and sequencing (DERBY-seq) to identify artificially enhanced and evolved reprogramming transcription factors. DERBY-seq entails pooled screens with libraries of positionally randomised genes, cell selection based on phenotypic readouts, and genotyping by amplicon sequencing for candidate identification. We benchmark this approach using pluripotency reprogramming with libraries based on the reprogramming factor SOX2 and the reprogramming incompetent endodermal factor SOX17. We identified several SOX2 variants outperforming the wild-type protein in three- and four-factor cocktails. The most effective variants were discovered from the SOX17 library, demonstrating that this factor can be converted into a highly potent inducer of pluripotency with a range of diverse modifications. We propose DERBY-seq as a broad-based approach to discover reprogramming factors for any donor/target cell combination applicable to direct lineage reprogramming in vitro and in vivo.


Asunto(s)
Diferenciación Celular , Reprogramación Celular/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Ingeniería de Proteínas , Animales , Sitios de Unión , Biomarcadores , Línea Celular , Biblioteca de Genes , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Motivos de Nucleótidos , Unión Proteica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
J Mol Biol ; 429(23): 3626-3634, 2017 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-29050852

RESUMEN

Sox2 and Pax6 co-regulate genes in neural lineages and the lens by forming a ternary complex likely facilitated allosterically through DNA. We used the quantitative and scalable cooperativity-by-sequencing (Coop-seq) approach to interrogate Sox2/Pax6 dimerization on a DNA library where five positions of the Pax6 half-site were randomized yielding 1024 cooperativity factors. Consensus positions normally required for the high-affinity DNA binding by Pax6 need to be mutated for effective dimerization with Sox2. Out of the five randomized bases, a 5' thymidine is present in most of the top ranking elements. However, this thymidine maps to a region outside of the Pax half site and is not expected to directly interact with Pax6 in known binding modes suggesting structural reconfigurations. Re-analysis of ChIP-seq data identified several genomic regions where the cooperativity promoting sequence pattern is co-bound by Sox2 and Pax6. A highly conserved Sox2/Pax6 bound site near the Sprouty2 locus was verified to promote cooperative dimerization designating Sprouty2 as a potential target reliant on Sox2/Pax6 cooperativity in several neural cell types. Collectively, the functional interplay of Sox2 and Pax6 demands the relaxation of high-affinity binding sites and is enabled by alternative DNA sequences. We conclude that this binding mode evolved to warrant that a subset of target genes is only regulated in the presence of suitable partner factors.


Asunto(s)
ADN/metabolismo , Factor de Transcripción PAX6/metabolismo , Factores de Transcripción SOXB1/metabolismo , Análisis de Secuencia de ADN/métodos , ADN/química , ADN/genética , Humanos , Modelos Moleculares , Factor de Transcripción PAX6/química , Factor de Transcripción PAX6/genética , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Factores de Transcripción SOXB1/química , Factores de Transcripción SOXB1/genética
9.
Structure ; 22(9): 1274-1286, 2014 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-25126959

RESUMEN

In pluripotent cells, OCT4 associates with SOX2 to maintain pluripotency or with SOX17 to induce primitive endoderm commitment. The OCT4-SOX2 and OCT4-SOX17 combinations bind mutually exclusive to two distinct composite DNA elements, known as the "canonical" and "compressed" motifs, respectively. The structural basis for the OCT4-SOX17 cooperativity is unknown. Whereas SOX17 has been engineered to replace SOX2 in the pluripotency circuitry, all generated SOX2 mutants have failed to act like SOX17. From molecular simulations, we revealed the OCT4-SOX17 interaction interface and elucidated the SOX-dependent motif preference of OCT4. Moreover, we designed a SOX2 mutant that we predicted and confirmed experimentally to bind cooperatively with OCT4 to the compressed motif. Ultimately, we found a strong correlation between the experimental and calculated relative cooperative-binding free energies of 12 OCT4-SOX-DNA complexes. Therefore, we validated the OCT4-SOX interfaces and demonstrated that in silico design of DNA-binding cooperativity is suitable for altering transcriptional circuitries.


Asunto(s)
Proteínas HMGB/química , Factor 3 de Transcripción de Unión a Octámeros/química , Factores de Transcripción SOXB1/química , Factores de Transcripción SOXF/química , Células Madre/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Diferenciación Celular , Secuencia de Consenso , ADN/química , Proteínas HMGB/genética , Interacciones Hidrofóbicas e Hidrofílicas , Ratones , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Factor 3 de Transcripción de Unión a Octámeros/genética , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXF/genética , Termodinámica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA