Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Metab Eng ; 60: 77-86, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32247827

RESUMEN

Maintaining a metabolic steady state is essential for an organism's fitness and survival when confronted with environmental stress, and metabolic imbalance can be reversed by exposing the organism to fasting. Here, we attempted to apply this physiological principle to mammalian cell cultures to improve cellular fitness and consequently their ability to express recombinant proteins. We showed that transient vitamin B5 deprivation, an essential cofactor of central cellular metabolism, can quickly and irreversibly affect mammalian cell growth and division. A selection method was designed that relies on mammalian cell dependence on vitamin B5 for energy production, using the co-expression of the B5 transporter SLC5A6 and a gene of interest. We demonstrated that vitamin B5 selection persistently activates peroxisome proliferator-activated receptors (PPAR), a family of transcription factors involved in energy homeostasis, thereby altering lipid metabolism, improving cell fitness and therapeutic protein production. Thus, stable PPAR activation may constitute a cellular memory of past deprivation state, providing increased resistance to further potential fasting events. In other words, our results imply that cultured cells, once exposed to metabolic starvation, may display an improved metabolic fitness as compared to non-exposed cells, allowing increased resistance to cellular stress.


Asunto(s)
Homeostasis , Ácido Pantoténico/deficiencia , Ácido Pantoténico/metabolismo , Proteínas Recombinantes/biosíntesis , Animales , Células CHO , División Celular , Células Cultivadas , Cricetinae , Cricetulus , Metabolismo Energético , Vectores Genéticos , Metabolismo de los Lípidos/fisiología , PPAR alfa/biosíntesis , PPAR alfa/genética , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Estrés Fisiológico , Simportadores/metabolismo
2.
Biotechnol Bioeng ; 117(12): 3628-3638, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32779734

RESUMEN

A clonally derived (or "monoclonal") cell line is a cell population derived from a single progenitor cell. Clonally derived cell lines are required for many biotechnological applications. For instance, recombinant mammalian cells used to produce therapeutic proteins are expected by regulatory authorities to be clonally derived. Assurance of clonal derivation (or "clonality") is usually obtained from the characterization of the procedure used for cell cloning, for instance by assessing the success rate of single-cell sorting but not by assessing the cell line itself. We have developed a method to assess clonal derivation directly from the genetic makeup of cells. The genomic test of clonality is based on whole-genome sequencing and statistical analysis of single nucleotide variants. This approach quantifies the clonal fractions present in nonclonal samples and it provides a measure of the probability that a cell line is derived from a single cell. Upon experimental validation of the test, we show that it is highly accurate and that it can robustly detect minor clonal fractions of as little as 1% of the cell population. Moreover, we find that it is applicable to various cell line development protocols. This approach can simplify development protocols and shorten timelines while ensuring clonal derivation with high confidence.


Asunto(s)
Células Clonales , Polimorfismo de Nucleótido Simple , Secuenciación Completa del Genoma , Animales , Productos Biológicos , Células CHO , Cricetulus
3.
Biotechnol Bioeng ; 117(4): 1101-1116, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31956982

RESUMEN

Despite extensive research conducted to increase protein production from Chinese hamster ovary (CHO) cells, cellular bottlenecks often remain, hindering high yields. In this study, a transcriptomic analysis led to the identification of 32 genes that are consistently upregulated in high producer clones and thus might mediate high productivity. Candidate genes were associated with functions such as signaling, protein folding, cytoskeleton organization, and cell survival. We focused on two engineering targets, Erp27, which binds unfolded proteins and the Erp57 disulfide isomerase in the endoplasmic reticulum, and Foxa1, a pioneering transcription factor involved in organ development. Erp27 moderate overexpression increased production of an easy-to-express antibody, whereas Erp27 and Erp57 co-overexpression increased cell density, viability, and the yield of difficult-to-express proteins. Foxa1 overexpression increased cell density, cell viability, and easy- and difficult-to-express protein yields, whereas it decreased reactive oxygen species late in fed-batch cultures. Foxa1 overexpression upregulated two other candidate genes that increased the production of difficult- and/or easy-to-express proteins, namely Ca3, involved in protecting cells from oxidative stress, and Tagap, involved in signaling and cytoskeleton remodeling. Overall, several genes allowing to overcome CHO cell bottlenecks were identified, including Foxa1, which mediated multiple favorable metabolic changes that improve therapeutic protein yields.


Asunto(s)
Ingeniería Celular/métodos , Factor Nuclear 3-alfa del Hepatocito , Proteínas Recombinantes , Animales , Células CHO , Supervivencia Celular , Cricetinae , Cricetulus , Factor Nuclear 3-alfa del Hepatocito/genética , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Pliegue de Proteína , Proteínas Recombinantes/análisis , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
4.
Biotechnol Bioeng ; 117(4): 1117-1126, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31956990

RESUMEN

In this study, we assessed the importance of cytoskeleton organization in the mammalian cells used to produce therapeutic proteins. Two cytoskeletal genes, Actin alpha cardiac muscle 1 (ACTC1) and a guanosine triphosphate GTPase-activating protein (TAGAP), were found to be upregulated in highly productive therapeutic protein-expressing Chinese hamster ovary (CHO) cells selected by the deprivation of vitamin B5. We report here that the overexpression of the ACTC1 protein was able to improve significantly recombinant therapeutic production, as well as to decrease the levels of toxic lactate metabolic by-products. ACTC1 overexpression was accompanied by altered as well as decreased polymerized actin, which was associated with high protein production by CHO cell cultured in suspension. We suggest that the depolymerization of actin and the possible modulation of integrin signaling, as well as changes in basal metabolism, may be driving the increase of protein secretion by CHO cells.


Asunto(s)
Citoesqueleto de Actina , Actinas , Proteínas Recombinantes , Citoesqueleto de Actina/química , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Proteínas Recombinantes/análisis , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
5.
Biotechnol Bioeng ; 117(2): 466-485, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31631325

RESUMEN

The Chinese hamster ovary (CHO) cells used to produce biopharmaceutical proteins are known to contain type-C endogenous retrovirus (ERV) sequences in their genome and to release retroviral-like particles. Although evidence for their infectivity is missing, this has raised safety concerns. As the genomic origin of these particles remained unclear, we characterized type-C ERV elements at the genome, transcriptome, and viral particle RNA levels. We identified 173 type-C ERV sequences clustering into three functionally conserved groups. Transcripts from one type-C ERV group were full-length, with intact open reading frames, and cognate viral genome RNA was loaded into retroviral-like particles, suggesting that this ERV group may produce functional viruses. CRISPR-Cas9 genome editing was used to disrupt the gag gene of the expressed type-C ERV group. Comparison of CRISPR-derived mutations at the DNA and RNA level led to the identification of a single ERV as the main source of the release of RNA-loaded viral particles. Clones bearing a Gag loss-of-function mutation in this ERV showed a reduction of RNA-containing viral particle release down to detection limits, without compromising cell growth or therapeutic protein production. Overall, our study provides a strategy to mitigate potential viral particle contaminations resulting from ERVs during biopharmaceutical manufacturing.


Asunto(s)
Células CHO/virología , Retrovirus Endógenos , Mutagénesis Sitio-Dirigida/métodos , ARN Viral , Virión/genética , Animales , Sistemas CRISPR-Cas , Cricetinae , Cricetulus , Contaminación de Medicamentos/prevención & control , Retrovirus Endógenos/genética , Retrovirus Endógenos/metabolismo , Edición Génica , Genoma Viral/genética , Mutación con Pérdida de Función/genética , ARN Viral/genética , ARN Viral/metabolismo
6.
Mol Ther ; 26(4): 1093-1108, 2018 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-29503200

RESUMEN

Duchenne muscular dystrophy (DMD) is a lethal muscle-wasting disease currently without cure. We investigated the use of the PiggyBac transposon for full-length dystrophin expression in murine mesoangioblast (MABs) progenitor cells. DMD murine MABs were transfected with transposable expression vectors for full-length dystrophin and transplanted intramuscularly or intra-arterially into mdx/SCID mice. Intra-arterial delivery indicated that the MABs could migrate to regenerating muscles to mediate dystrophin expression. Intramuscular transplantation yielded dystrophin expression in 11%-44% of myofibers in murine muscles, which remained stable for the assessed period of 5 months. The satellite cells isolated from transplanted muscles comprised a fraction of MAB-derived cells, indicating that the transfected MABs may colonize the satellite stem cell niche. Transposon integration site mapping by whole-genome sequencing indicated that 70% of the integrations were intergenic, while none was observed in an exon. Muscle resistance assessment by atomic force microscopy indicated that 80% of fibers showed elasticity properties restored to those of wild-type muscles. As measured in vivo, transplanted muscles became more resistant to fatigue. This study thus provides a proof-of-principle that PiggyBac transposon vectors may mediate full-length dystrophin expression as well as functional amelioration of the dystrophic muscles within a potential autologous cell-based therapeutic approach of DMD.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Elementos Transponibles de ADN , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Distrofia Muscular de Duchenne/genética , Mioblastos/metabolismo , Mioblastos/trasplante , Animales , Línea Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Modelos Animales de Enfermedad , Distrofina/genética , Técnica del Anticuerpo Fluorescente , Dosificación de Gen , Expresión Génica , Orden Génico , Genes Reporteros , Masculino , Ratones , Ratones Endogámicos mdx , Ratones SCID , Distrofia Muscular de Duchenne/patología , Distrofia Muscular de Duchenne/fisiopatología , Distrofia Muscular de Duchenne/terapia , Fenotipo , Transgenes , Trasplante Autólogo
7.
Nucleic Acids Res ; 44(6): e56, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-26657630

RESUMEN

DNA double stranded breaks (DSBs) are one of the most deleterious types of DNA lesions. The main pathways responsible for repairing these breaks in eukaryotic cells are homologous recombination (HR) and non-homologous end-joining (NHEJ). However, a third group of still poorly characterized DSB repair pathways, collectively termed microhomology-mediated end-joining (MMEJ), relies on short homologies for the end-joining process. Here, we constructed GFP reporter assays to characterize and distinguish MMEJ variant pathways, namely the simple MMEJ and the DNA synthesis-dependent (SD)-MMEJ mechanisms. Transfection of these assay vectors in Chinese hamster ovary (CHO) cells and characterization of the repaired DNA sequences indicated that while simple MMEJ is able to mediate relatively efficient DSB repair if longer microhomologies are present, the majority of DSBs were repaired using the highly error-prone SD-MMEJ pathway. To validate the involvement of DNA synthesis in the repair process, siRNA knock-down of different genes proposed to play a role in MMEJ were performed, revealing that the knock-down of DNA polymerase θ inhibited DNA end resection and repair through simple MMEJ, thus favoring the other repair pathway. Overall, we conclude that this approach provides a convenient assay to study MMEJ-related DNA repair pathways.


Asunto(s)
Bioensayo , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , ADN Polimerasa Dirigida por ADN/genética , ADN/metabolismo , Animales , Secuencia de Bases , Células CHO , Cricetulus , ADN/química , ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Recombinación Homóloga , Datos de Secuencia Molecular , Plásmidos/química , Plásmidos/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Homología de Secuencia de Ácido Nucleico , Transfección , ADN Polimerasa theta
8.
Biotechnol Bioeng ; 114(8): 1791-1802, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28214330

RESUMEN

We developed a method for the fast sorting and selection of mammalian cells expressing and secreting a protein at high levels. This procedure relies on cell capture using an automated microfluidic device handling antibody-coupled magnetic microparticles and on a timed release of the cells from the microparticles after capture. Using clinically compatible materials and procedures, we show that this approach is able to discriminate between cells that truly secrete high amounts of a protein from those that just display it at high levels on their surface without properly releasing it. When coupled to a cell colony imaging and picking device, this approach allowed the identification of CHO cell clones secreting a therapeutic protein at high levels that were not achievable without the cell sorting procedure. Biotechnol. Bioeng. 2017;114: 1791-1802. © 2017 Wiley Periodicals, Inc.


Asunto(s)
Células CHO/citología , Células CHO/metabolismo , Separación Celular/métodos , Nanopartículas de Magnetita/química , Proteínas Recombinantes/metabolismo , Animales , Células CHO/efectos de la radiación , Cricetulus , Nanopartículas de Magnetita/efectos de la radiación , Coloración y Etiquetado/métodos
9.
Biotechnol Bioeng ; 114(2): 384-396, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27575535

RESUMEN

Untargeted plasmid integration into mammalian cell genomes remains a poorly understood and inefficient process. The formation of plasmid concatemers and their genomic integration has been ascribed either to non-homologous end-joining (NHEJ) or homologous recombination (HR) DNA repair pathways. However, a direct involvement of these pathways has remained unclear. Here, we show that the silencing of many HR factors enhanced plasmid concatemer formation and stable expression of the gene of interest in Chinese hamster ovary (CHO) cells, while the inhibition of NHEJ had no effect. However, genomic integration was decreased by the silencing of specific HR components, such as Rad51, and DNA synthesis-dependent microhomology-mediated end-joining (SD-MMEJ) activities. Genome-wide analysis of the integration loci and junction sequences validated the prevalent use of the SD-MMEJ pathway for transgene integration close to cellular genes, an effect shared with matrix attachment region (MAR) DNA elements that stimulate plasmid integration and expression. Overall, we conclude that SD-MMEJ is the main mechanism driving the illegitimate genomic integration of foreign DNA in CHO cells, and we provide a recombination engineering approach that increases transgene integration and recombinant protein expression in these cells. Biotechnol. Bioeng. 2017;114: 384-396. © 2016 The Authors. Biotechnology and Bioengineering published by Wiley Periodicals, Inc.


Asunto(s)
Cromatina/genética , Ingeniería Genética/métodos , Regiones de Fijación a la Matriz/genética , Proteínas Recombinantes/genética , Recombinación Genética/genética , Animales , Anticuerpos/química , Anticuerpos/genética , Anticuerpos/metabolismo , Células CHO , Cricetinae , Cricetulus , Técnicas de Silenciamiento del Gen , Humanos , Plásmidos/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Transgenes/genética
10.
J Cell Sci ; 127(Pt 15): 3240-56, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24895400

RESUMEN

Here, we identify a role for the matrilin-2 (Matn2) extracellular matrix protein in controlling the early stages of myogenic differentiation. We observed Matn2 deposition around proliferating, differentiating and fusing myoblasts in culture and during muscle regeneration in vivo. Silencing of Matn2 delayed the expression of the Cdk inhibitor p21 and of the myogenic genes Nfix, MyoD and Myog, explaining the retarded cell cycle exit and myoblast differentiation. Rescue of Matn2 expression restored differentiation and the expression of p21 and of the myogenic genes. TGF-ß1 inhibited myogenic differentiation at least in part by repressing Matn2 expression, which inhibited the onset of a positive-feedback loop whereby Matn2 and Nfix activate the expression of one another and activate myoblast differentiation. In vivo, myoblast cell cycle arrest and muscle regeneration was delayed in Matn2(-/-) relative to wild-type mice. The expression levels of Trf3 and myogenic genes were robustly reduced in Matn2(-/-) fetal limbs and in differentiating primary myoblast cultures, establishing Matn2 as a key modulator of the regulatory cascade that initiates terminal myogenic differentiation. Our data thus identify Matn2 as a crucial component of a genetic switch that modulates the onset of tissue repair.


Asunto(s)
Matriz Extracelular/metabolismo , Proteínas Matrilinas/metabolismo , Músculos/fisiología , Mioblastos/fisiología , Necrosis/terapia , Animales , Apoptosis/genética , Línea Celular , Proliferación Celular/genética , Venenos Elapídicos/administración & dosificación , Humanos , Proteínas Matrilinas/genética , Ratones , Ratones Noqueados , Desarrollo de Músculos/genética , Músculos/patología , Necrosis/inducido químicamente , Ratas , Ratas Wistar , Regeneración/genética , Factores de Tiempo
11.
Nucleic Acids Res ; 42(1): 193-204, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24071586

RESUMEN

In eukaryotic cells, transgene expression levels may be limited by an unfavourable chromatin structure at the integration site. Epigenetic regulators are DNA sequences which may protect transgenes from such position effect. We evaluated different epigenetic regulators for their ability to protect transgene expression at telomeres, which are commonly associated to low or inconsistent expression because of their repressive chromatin environment. Although to variable extents, matrix attachment regions (MARs), ubiquitous chromatin opening element (UCOE) and the chicken cHS4 insulator acted as barrier elements, protecting a telomeric-distal transgene from silencing. MARs also increased the probability of silent gene reactivation in time-course experiments. Additionally, all MARs improved the level of expression in non-silenced cells, unlike other elements. MARs were associated to histone marks usually linked to actively expressed genes, especially acetylation of histone H3 and H4, suggesting that they may prevent the spread of silencing chromatin by imposing acetylation marks on nearby nucleosomes. Alternatively, an UCOE was found to act by preventing deposition of repressive chromatin marks. We conclude that epigenetic DNA elements used to enhance and stabilize transgene expression all have specific epigenetic signature that might be at the basis of their mode of action.


Asunto(s)
ADN/química , Epistasis Genética , Silenciador del Gen , Histonas/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Telómero , Cromatina/metabolismo , Células HeLa , Humanos , Regiones de Fijación a la Matriz , Transgenes
12.
Liver Int ; 35(4): 1185-94, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25293436

RESUMEN

BACKGROUND & AIMS: Knockout studies of the murine Nuclear Factor I-C (NFI-C) transcription factor revealed abnormal skin wound healing and growth of its appendages, suggesting a role in controlling cell proliferation in adult regenerative processes. Liver regeneration following partial hepatectomy (PH) is a well-established regenerative model whereby changes elicited in hepatocytes lead to their rapid and phased proliferation. Although NFI-C is highly expressed in the liver, no hepatic function was yet established for this transcription factor. This study aimed to determine whether NFI-C may play a role in hepatocyte proliferation and liver regeneration. METHODS: Liver regeneration and cell proliferation pathways following two-thirds PH were investigated in NFI-C knockout (ko) and wild-type (wt) mice. RESULTS: We show that the absence of NFI-C impaired hepatocyte proliferation because of plasminogen activator I (PAI-1) overexpression and the subsequent suppression of urokinase plasminogen activator (uPA) activity and hepatocyte growth factor (HGF) signalling, a potent hepatocyte mitogen. This indicated that NFI-C first acts to promote hepatocyte proliferation at the onset of liver regeneration in wt mice. The subsequent transient down regulation of NFI-C, as can be explained by a self-regulatory feedback loop with transforming growth factor beta 1 (TGF-ß1), may limit the number of hepatocytes entering the first wave of cell division and/or prevent late initiations of mitosis. CONCLUSION: NFI-C acts as a regulator of the phased hepatocyte proliferation during liver regeneration.


Asunto(s)
Proliferación Celular , Regeneración Hepática , Hígado/metabolismo , Factores de Transcripción NFI/metabolismo , Animales , Retroalimentación Fisiológica , Regulación de la Expresión Génica , Genotipo , Hepatectomía/métodos , Factor de Crecimiento de Hepatocito/genética , Factor de Crecimiento de Hepatocito/metabolismo , Hígado/patología , Hígado/fisiopatología , Hígado/cirugía , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción NFI/deficiencia , Factores de Transcripción NFI/genética , Fenotipo , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Transducción de Señal , Factores de Tiempo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Activador de Plasminógeno de Tipo Uroquinasa/genética , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo
13.
Metab Eng ; 21: 91-102, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23380542

RESUMEN

The ability to efficiently produce recombinant proteins in a secreted form is highly desirable and cultured mammalian cells such as CHO cells have become the preferred host as they secrete proteins with human-like post-translational modifications. However, attempts to express high levels of particular proteins in CHO cells may consistently result in low yields, even for non-engineered proteins such as immunoglobulins. In this study, we identified the responsible faulty step at the stage of translational arrest, translocation and early processing for such a "difficult-to-express" immunoglobulin, resulting in improper cleavage of the light chain and its precipitation in an insoluble cellular fraction unable to contribute to immunoglobulin assembly. We further show that proper processing and secretion were restored by over-expressing human signal receptor protein SRP14 and other components of the secretion pathway. This allowed the expression of the difficult-to-express protein to high yields, and it also increased the production of an easy-to-express protein. Our results demonstrate that components of the secretory and processing pathways can be limiting, and that engineering of the secretory pathway may be used to improve the secretion efficiency of therapeutic proteins from CHO cells.


Asunto(s)
Ingeniería Genética , Vías Secretoras/genética , Partícula de Reconocimiento de Señal/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Humanos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Partícula de Reconocimiento de Señal/genética
14.
BMC Genomics ; 14: 99, 2013 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-23402308

RESUMEN

BACKGROUND: The Nuclear Factor I (NFI) family of DNA binding proteins (also called CCAAT box transcription factors or CTF) is involved in both DNA replication and gene expression regulation. Using chromatin immuno-precipitation and high throughput sequencing (ChIP-Seq), we performed a genome-wide mapping of NFI DNA binding sites in primary mouse embryonic fibroblasts. RESULTS: We found that in vivo and in vitro NFI DNA binding specificities are indistinguishable, as in vivo ChIP-Seq NFI binding sites matched predictions based on previously established position weight matrix models of its in vitro binding specificity. Combining ChIP-Seq with mRNA profiling data, we found that NFI preferentially associates with highly expressed genes that it up-regulates, while binding sites were under-represented at expressed but unregulated genes. Genomic binding also correlated with markers of transcribed genes such as histone modifications H3K4me3 and H3K36me3, even outside of annotated transcribed loci, implying NFI in the control of the deposition of these modifications. Positional correlation between + and - strand ChIP-Seq tags revealed that, in contrast to other transcription factors, NFI associates with a nucleosomal length of cleavage-resistant DNA, suggesting an interaction with positioned nucleosomes. In addition, NFI binding prominently occurred at boundaries displaying discontinuities in histone modifications specific of expressed and silent chromatin, such as loci submitted to parental allele-specific imprinted expression. CONCLUSIONS: Our data thus suggest that NFI nucleosomal interaction may contribute to the partitioning of distinct chromatin domains and to epigenetic gene expression regulation.NFI ChIP-Seq and input control DNA data were deposited at Gene Expression Omnibus (GEO) repository under accession number GSE15844. Gene expression microarray data for mouse embryonic fibroblasts are on GEO accession number GSE15871.


Asunto(s)
Epigénesis Genética , Factores de Transcripción NFI/genética , Nucleosomas/genética , Regiones Promotoras Genéticas , Animales , Sitios de Unión , Cromatina/genética , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Mapeo Cromosómico , Metilación de ADN , Replicación del ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Genoma , Ratones , Factores de Transcripción NFI/metabolismo , Nucleosomas/metabolismo , Activación Transcripcional/genética
15.
BMC Mol Biol ; 14: 26, 2013 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-24295286

RESUMEN

BACKGROUND: The in vivo transfer of naked plasmid DNA into organs such as muscles is commonly used to assess the expression of prophylactic or therapeutic genes in animal disease models. RESULTS: In this study, we devised vectors allowing a tight regulation of transgene expression in mice from such non-viral vectors using a doxycycline-controlled network of activator and repressor proteins. Using these vectors, we demonstrate proper physiological response as consequence of the induced expression of two therapeutically relevant proteins, namely erythropoietin and utrophin. Kinetic studies showed that the induction of transgene expression was only transient, unless epigenetic regulatory elements termed Matrix Attachment Regions, or MAR, were inserted upstream of the regulated promoters. Using episomal plasmid rescue and quantitative PCR assays, we observed that similar amounts of plasmids remained in muscles after electrotransfer with or without MAR elements, but that a significant portion had integrated into the muscle fiber chromosomes. Interestingly, the MAR elements were found to promote plasmid genomic integration but to oppose silencing effects in vivo, thereby mediating long-term expression. CONCLUSIONS: This study thus elucidates some of the determinants of transient or sustained expression from the use of non-viral regulated vectors in vivo.


Asunto(s)
Regulación de la Expresión Génica , Técnicas de Transferencia de Gen , Vectores Genéticos , Regiones de Fijación a la Matriz , Plásmidos/genética , Transgenes , Animales , Doxiciclina/farmacología , Epigénesis Genética , Eritropoyetina/genética , Eritropoyetina/metabolismo , Femenino , Humanos , Cinética , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Músculos/metabolismo , Utrofina/genética , Utrofina/metabolismo
16.
Biotechnol Bioeng ; 110(4): 1153-63, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23096947

RESUMEN

The generation of a high productivity cell line is a critical step in the production of a therapeutic protein. Many innovative engineering strategies have been devised in order to maximize the expression rate of production cells for increased process efficiency. Less effort has focused on improvements to the cell line generation process, which is typically long and laborious when using mammalian cells. Based on unexpected findings when generating stable CHO cell lines expressing human IL-17F, we studied the benefit of expressing this protein during the establishment of production cell lines. We demonstrate that IL-17F expression enhances the rate of selection and overall number of selected cell lines as well as their transgene expression levels. We also show that this benefit is observed with different parental CHO cell lines and selection systems. Furthermore, IL-17F expression improves the efficiency of cell line subcloning processes. IL-17F can therefore be exploited in a standard manufacturing process to obtain higher productivity clones in a reduced time frame.


Asunto(s)
División Celular , Interleucina-17/genética , Animales , Células CHO , Cricetinae , Cricetulus , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Vectores Genéticos , Humanos , Proteínas Recombinantes/biosíntesis
17.
Nucleic Acids Res ; 39(15): e104, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21652640

RESUMEN

Gene transfer and expression in eukaryotes is often limited by a number of stably maintained gene copies and by epigenetic silencing effects. Silencing may be limited by the use of epigenetic regulatory sequences such as matrix attachment regions (MAR). Here, we show that successive transfections of MAR-containing vectors allow a synergistic increase of transgene expression. This finding is partly explained by an increased entry into the cell nuclei and genomic integration of the DNA, an effect that requires both the MAR element and iterative transfections. Fluorescence in situ hybridization analysis often showed single integration events, indicating that DNAs introduced in successive transfections could recombine. High expression was also linked to the cell division cycle, so that nuclear transport of the DNA occurs when homologous recombination is most active. Use of cells deficient in either non-homologous end-joining or homologous recombination suggested that efficient integration and expression may require homologous recombination-based genomic integration of MAR-containing plasmids and the lack of epigenetic silencing events associated with tandem gene copies. We conclude that MAR elements may promote homologous recombination, and that cells and vectors can be engineered to take advantage of this property to mediate highly efficient gene transfer and expression.


Asunto(s)
Regiones de Fijación a la Matriz , Recombinación Genética , Transfección , Transgenes , Transporte Activo de Núcleo Celular , Animales , Ciclo Celular , Línea Celular , Núcleo Celular/metabolismo , ADN/química , ADN/metabolismo , Dosificación de Gen , Expresión Génica , Vectores Genéticos , Plásmidos/genética , Homología de Secuencia de Ácido Nucleico
18.
J Biol Chem ; 285(44): 34115-25, 2010 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-20729551

RESUMEN

Skin appendages such as teeth and hair share several common signaling pathways. The nuclear factor I C (NFI-C) transcription factor has been implicated in tooth development, but a potential role in hair growth had not been assessed. In this study we found that NFI-C regulates the onset of the hair growth cycle. NFI-C(-/-) mice were delayed in the transition from the telogen to anagen phase of the hair follicle cycle after either experimental depilation or spontaneous hair loss. Lack of NFI-C resulted in delayed induction of the sonic hedgehog, Wnt5a, and Lef1 gene expression, which are key regulators of the hair follicle growth initiation. NFI-C(-/-) mice also showed elevated levels of transforming growth factor ß1 (TGF-ß1), an inhibitor of keratinocyte proliferation, and of the cell cycle inhibitor p21 at telogen. Reduced expression of Ki67, a marker of cell proliferation, was noted at the onset of anagen, indicating impaired activation of the hair progenitor cells. These findings implicate NFI-C in the repression of TGF-ß1 signaling during telogen stage, resulting in the delay of progenitor cell proliferation and hair follicle regeneration in NFI-C-deficient mice. Taken together with prior observations, these findings also designate NFI-C as a regulator of adult progenitor cell proliferation and of postnatal tissue growth or regeneration.


Asunto(s)
Folículo Piloso/metabolismo , Factores de Transcripción NFI/química , Factor de Crecimiento Transformador beta/metabolismo , Animales , Proliferación Celular , Cabello/fisiología , Inmunohistoquímica/métodos , Factor de Unión 1 al Potenciador Linfoide/biosíntesis , Ratones , Ratones Noqueados , Ratones Transgénicos , Fenotipo , Transducción de Señal , Células Madre/citología , Proteínas Wnt/biosíntesis , Proteína Wnt-5a
19.
BMC Genomics ; 12: 181, 2011 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-21473784

RESUMEN

BACKGROUND: Multiplex experimental assays coupled to computational predictions are being increasingly employed for the simultaneous analysis of many specimens at the genome scale, which quickly generates very large amounts of data. However, inferring valuable biological information from the comparisons of very large genomic datasets still represents an enormous challenge. RESULTS: As a study model, we chose the NFI/CTF family of mammalian transcription factors and we compared the results obtained from a genome-wide study of its binding sites with chromatin structure assays, gene expression microarray data, and in silico binding site predictions. We found that NFI/CTF family members preferentially bind their DNA target sites when they are located around transcription start sites when compared to control datasets generated from the random subsampling of the complete set of NFI binding sites. NFI proteins preferably associate with the upstream regions of genes that are highly expressed and that are enriched in active chromatin modifications such as H3K4me3 and H3K36me3. We postulate that this is a causal association and that NFI proteins mainly act as activators of transcription. This was documented for one member of the family (NFI-C), which revealed as a more potent gene activator than repressor in global gene expression analysis. Interestingly, we also discovered the association of NFI with the tri-methylation of lysine 9 of histone H3, a chromatin marker previously associated with the protection against silencing of telomeric genes by NFI. CONCLUSION: Taken together, we illustrate approaches that can be taken to analyze large genomic data, and provide evidence that NFI family members may act in conjunction with specific chromatin modifications to activate gene expression.


Asunto(s)
Factores de Transcripción NFI/genética , Regiones Promotoras Genéticas , Factores de Transcripción/genética , Activación Transcripcional , Algoritmos , Animales , Análisis por Conglomerados , Regulación de la Expresión Génica , Genómica/métodos , Histonas/metabolismo , Metilación , Ratones , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Unión Proteica , Sitio de Iniciación de la Transcripción
20.
J Biotechnol ; 341: 103-112, 2021 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-34560160

RESUMEN

Integrative non-viral vectors such as transposons engineered to mediate targeted gene transfer into safe harbor sites in the genome may be a promising approach for the production of therapeutic proteins or for gene therapy in an efficient and secure way. In this context, we designed and evaluated two strategies for targeting the nuclear ribosomal DNA (rDNA) loci. One approach relied on the co-location of the transposase and transposon near transcriptionally active rDNA copies using a nucleolar localization signal (NoLS). Another one consisted of targeting the 18S-coding region in the rDNA loci using a NoLS-FokI-dCas9 endonuclease to perform targeted transgene knock-in. We show that integration into the rDNA of Chinese hamster ovary (CHO) cells can be achieved at a high frequency using the piggyBac transposon system, indicating that the rDNA is highly accessible for transposition. Consistently, rDNA-targeted transposition events were most frequently obtained when both the piggyBac transposon DNA and the transposase were nucleoli-targeted, yielding cells displaying stable and homogeneous expression of the transgene. This approach thus provides an alternative strategy to improve targeted transgene delivery and protein expression using CHO cells.


Asunto(s)
Elementos Transponibles de ADN , Transposasas , Animales , Células CHO , Cricetinae , Cricetulus , Elementos Transponibles de ADN/genética , ADN Ribosómico , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos , Transposasas/genética , Transposasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA