Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35046049

ABSTRACT

Cancer immunotherapy frequently fails because most carcinomas have few T cells, suggesting that cancers can suppress T cell infiltration. Here, we show that cancer cells of human pancreatic ductal adenocarcinoma (PDA), colorectal cancer, and breast cancer are coated with transglutaminase-2 (TGM2)-dependent covalent CXCL12-keratin-19 (KRT19) heterodimers that are organized as filamentous networks. Since a dimeric form of CXCL12 suppresses the motility of human T cells, we determined whether this polymeric CXCL12-KRT19 coating mediated T cell exclusion. Mouse tumors containing control PDA cells exhibited the CXCL12-KRT19 coating, excluded T cells, and did not respond to treatment with anti-PD-1 antibody. Tumors containing PDA cells not expressing either KRT19 or TGM2 lacked the CXCL12-KRT19 coating, were infiltrated with activated CD8+ T cells, and growth was suppressed with anti-PD-1 antibody treatment. Thus, carcinomas assemble a CXCL12-KRT19 coating to evade cancer immune attack.


Subject(s)
Carcinoma/etiology , Carcinoma/metabolism , Chemokine CXCL12/metabolism , Cytotoxicity, Immunologic , Keratin-19/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Breast Neoplasms , Carcinoma/pathology , Cell Line, Tumor , Chemokine CXCL12/chemistry , Female , Humans , Keratin-19/chemistry , Male , Mice , Microsatellite Repeats , Pancreatic Neoplasms , Protein Binding , Protein Multimerization , Pancreatic Neoplasms
2.
Proc Natl Acad Sci U S A ; 117(46): 28960-28970, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33127761

ABSTRACT

Inhibition of the chemokine receptor CXCR4 in combination with blockade of the PD-1/PD-L1 T cell checkpoint induces T cell infiltration and anticancer responses in murine and human pancreatic cancer. Here we elucidate the mechanism by which CXCR4 inhibition affects the tumor immune microenvironment. In human immune cell-based chemotaxis assays, we find that CXCL12-stimulated CXCR4 inhibits the directed migration mediated by CXCR1, CXCR3, CXCR5, CXCR6, and CCR2, respectively, chemokine receptors expressed by all of the immune cell types that participate in an integrated immune response. Inhibiting CXCR4 in an experimental cancer medicine study by 1-wk continuous infusion of the small-molecule inhibitor AMD3100 (plerixafor) induces an integrated immune response that is detected by transcriptional analysis of paired biopsies of metastases from patients with microsatellite stable colorectal and pancreatic cancer. This integrated immune response occurs in three other examples of immune-mediated damage to noninfected tissues: Rejecting renal allografts, melanomas clinically responding to anti-PD1 antibody therapy, and microsatellite instable colorectal cancers. Thus, signaling by CXCR4 causes immune suppression in human pancreatic ductal adenocarcinoma and colorectal cancer by impairing the function of the chemokine receptors that mediate the intratumoral accumulation of immune cells.


Subject(s)
Colorectal Neoplasms/metabolism , Immunity/immunology , Pancreas/metabolism , Pancreatic Neoplasms/metabolism , Receptors, CXCR4/drug effects , Receptors, CXCR4/metabolism , Aged , Benzylamines , Carcinoma, Pancreatic Ductal , Chemokine CXCL12 , Colorectal Neoplasms/pathology , Cyclams , Female , Heterocyclic Compounds/antagonists & inhibitors , Humans , Immunotherapy , Male , Middle Aged , Pancreatic Neoplasms/pathology , Receptors, CCR2/metabolism , Receptors, CXCR3/metabolism , Receptors, CXCR5/metabolism , Receptors, CXCR6/metabolism , Receptors, Interleukin-8A/metabolism , Signal Transduction/drug effects , Tumor Microenvironment/immunology , Pancreatic Neoplasms
3.
Gastroenterology ; 158(6): 1682-1697.e1, 2020 05.
Article in English | MEDLINE | ID: mdl-32032585

ABSTRACT

BACKGROUND & AIMS: Esophageal adenocarcinomas (EACs) are heterogeneous and often preceded by Barrett's esophagus (BE). Many genomic changes have been associated with development of BE and EAC, but little is known about epigenetic alterations. We performed epigenetic analyses of BE and EAC tissues and combined these data with transcriptome and genomic data to identify mechanisms that control gene expression and genome integrity. METHODS: In a retrospective cohort study, we collected tissue samples and clinical data from 150 BE and 285 EAC cases from the Oesophageal Cancer Classification and Molecular Stratification consortium in the United Kingdom. We analyzed methylation profiles of all BE and EAC tissues and assigned them to subgroups using non-negative matrix factorization with k-means clustering. Data from whole-genome sequencing and transcriptome studies were then incorporated; we performed integrative methylation and RNA-sequencing analyses to identify genes that were suppressed with increased methylation in promoter regions. Levels of different immune cell types were computed using single-sample gene set enrichment methods. We derived 8 organoids from 8 EAC tissues and tested their sensitivity to different drugs. RESULTS: BE and EAC samples shared genome-wide methylation features, compared with normal tissues (esophageal, gastric, and duodenum; controls) from the same patients and grouped into 4 subtypes. Subtype 1 was characterized by DNA hypermethylation with a high mutation burden and multiple mutations in genes in cell cycle and receptor tyrosine signaling pathways. Subtype 2 was characterized by a gene expression pattern associated with metabolic processes (ATP synthesis and fatty acid oxidation) and lack methylation at specific binding sites for transcription factors; 83% of samples of this subtype were BE and 17% were EAC. The third subtype did not have changes in methylation pattern, compared with control tissue, but had a gene expression pattern that indicated immune cell infiltration; this tumor type was associated with the shortest time of patient survival. The fourth subtype was characterized by DNA hypomethylation associated with structure rearrangements, copy number alterations, with preferential amplification of CCNE1 (cells with this gene amplification have been reported to be sensitive to CDK2 inhibitors). Organoids with reduced levels of MGMT and CHFR expression were sensitive to temozolomide and taxane drugs. CONCLUSIONS: In a comprehensive integrated analysis of methylation, transcriptome, and genome profiles of more than 400 BE and EAC tissues, along with clinical data, we identified 4 subtypes that were associated with patient outcomes and potential responses to therapy.


Subject(s)
Adenocarcinoma/genetics , Barrett Esophagus/genetics , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Esophageal Mucosa/pathology , Esophageal Neoplasms/genetics , Adenocarcinoma/pathology , Aged , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Barrett Esophagus/drug therapy , Barrett Esophagus/pathology , Cyclin E/genetics , DNA Methylation/drug effects , Disease Progression , Epigenesis, Genetic/drug effects , Esophageal Neoplasms/pathology , Female , Gene Amplification , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Middle Aged , Oncogene Proteins/genetics , Promoter Regions, Genetic/genetics , RNA-Seq , Retrospective Studies , Temozolomide/pharmacology , Temozolomide/therapeutic use , Whole Genome Sequencing
4.
Gut ; 68(8): 1386-1395, 2019 08.
Article in English | MEDLINE | ID: mdl-31030191

ABSTRACT

OBJECTIVE: We have previously described a prognostic transcriptional signature in CD8 T cells that separates patients with IBD into two phenotypically distinct subgroups, termed IBD1 and IBD2. Here we sought to develop a blood-based test that could identify these subgroups without cell separation, and thus be suitable for clinical use in Crohn's disease (CD) and ulcerative colitis (UC). DESIGN: Patients with active IBD were recruited before treatment. Transcriptomic analyses were performed on purified CD8 T cells and/or whole blood. Phenotype data were collected prospectively. IBD1/IBD2 patient subgroups were identified by consensus clustering of CD8 T cell transcriptomes. In a training cohort, machine learning was used to identify groups of genes ('classifiers') whose differential expression in whole blood recreated the IBD1/IBD2 subgroups. Genes from the best classifiers were quantitative (q)PCR optimised, and further machine learning was used to identify the optimal qPCR classifier, which was locked down for further testing. Independent validation was sought in separate cohorts of patients with CD (n=66) and UC (n=57). RESULTS: In both validation cohorts, a 17-gene qPCR-based classifier stratified patients into two distinct subgroups. Irrespective of the underlying diagnosis, IBDhi patients (analogous to the poor prognosis IBD1 subgroup) experienced significantly more aggressive disease than IBDlo patients (analogous to IBD2), with earlier need for treatment escalation (hazard ratio=2.65 (CD), 3.12 (UC)) and more escalations over time (for multiple escalations within 18 months: sensitivity=72.7% (CD), 100% (UC); negative predictive value=90.9% (CD), 100% (UC)). CONCLUSION: This is the first validated prognostic biomarker that can predict prognosis in newly diagnosed patients with IBD and represents a step towards personalised therapy.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Colitis, Ulcerative , Crohn Disease , Adult , Biomarkers/blood , Colitis, Ulcerative/blood , Colitis, Ulcerative/diagnosis , Crohn Disease/blood , Crohn Disease/diagnosis , Diagnosis, Differential , Female , Gene Expression , Gene Expression Profiling/methods , Humans , Machine Learning , Male , Middle Aged , Phenotype , Predictive Value of Tests , Prognosis , Reproducibility of Results , Sensitivity and Specificity , Severity of Illness Index
5.
Sci Rep ; 8(1): 11847, 2018 08 07.
Article in English | MEDLINE | ID: mdl-30087403

ABSTRACT

Abnormal hippocampal neural plasticity has been implicated in behavioural abnormalities and complex neuropsychiatric conditions, including bipolar disorder (BD). However, the determinants of this neural alteration remain unknown. This work tests the hypothesis that the neurotransmitter serotonin (5-HT) is a key determinant of hippocampal neuroplasticity, and its absence leads to maladaptive behaviour relevant for BD. Depletion of brain 5-HT in Tph2 mutant mice resulted in reduced behavioural despair, reduced anxiety, marked aggression and lower habituation in novel environments, reminiscent of bipolar-associated manic behaviour. Treatment with valproate produced a substantial improvement of the mania-like behavioural phenotypes displayed by Tph2 mutants. Brain-wide fMRI mapping in mutants revealed functional hippocampal hyperactivity in which we also observed dramatically increased neuroplasticity. Importantly, remarkable correspondence between the transcriptomic profile of the Tph2 mutant hippocampus and neurons from bipolar disorder patients was observed. Chronic stress reversed the emotional phenotype and the hippocampal transcriptional landscape of Tph2 mutants. These changes were associated with inappropriate activation of transcriptional adaptive response to stress as assessed by gene set enrichment analyses in the hippocampus of Tph2 mutant mice. These findings delineate 5-HT as a critical determinant in BD associated maladaptive emotional responses and aberrant hippocampal neuroplasticity, and support the use of Tph2-/- mice as a new research tool for mechanistic and therapeutic research in bipolar disorder.


Subject(s)
Bipolar Disorder/prevention & control , Hippocampus/drug effects , Neuronal Plasticity/drug effects , Serotonin/metabolism , Tryptophan Hydroxylase/metabolism , Valproic Acid/pharmacology , Animals , Anticonvulsants/pharmacology , Anxiety/genetics , Anxiety/physiopathology , Anxiety/prevention & control , Bipolar Disorder/genetics , Bipolar Disorder/physiopathology , Brain/diagnostic imaging , Brain/drug effects , Brain/metabolism , Gene Expression Profiling/methods , Hippocampus/metabolism , Hippocampus/physiopathology , Magnetic Resonance Imaging/methods , Male , Mice, Inbred C57BL , Mice, Knockout , Neuronal Plasticity/genetics , Neurons/drug effects , Neurons/metabolism , Tryptophan Hydroxylase/genetics
6.
J Allergy Clin Immunol ; 142(4): 1285-1296, 2018 10.
Article in English | MEDLINE | ID: mdl-29477724

ABSTRACT

BACKGROUND: The genetic cause of primary immunodeficiency disease (PID) carries prognostic information. OBJECTIVE: We conducted a whole-genome sequencing study assessing a large proportion of the NIHR BioResource-Rare Diseases cohort. METHODS: In the predominantly European study population of principally sporadic unrelated PID cases (n = 846), a novel Bayesian method identified nuclear factor κB subunit 1 (NFKB1) as one of the genes most strongly associated with PID, and the association was explained by 16 novel heterozygous truncating, missense, and gene deletion variants. This accounted for 4% of common variable immunodeficiency (CVID) cases (n = 390) in the cohort. Amino acid substitutions predicted to be pathogenic were assessed by means of analysis of structural protein data. Immunophenotyping, immunoblotting, and ex vivo stimulation of lymphocytes determined the functional effects of these variants. Detailed clinical and pedigree information was collected for genotype-phenotype cosegregation analyses. RESULTS: Both sporadic and familial cases demonstrated evidence of the noninfective complications of CVID, including massive lymphadenopathy (24%), unexplained splenomegaly (48%), and autoimmune disease (48%), features prior studies correlated with worse clinical prognosis. Although partial penetrance of clinical symptoms was noted in certain pedigrees, all carriers have a deficiency in B-lymphocyte differentiation. Detailed assessment of B-lymphocyte numbers, phenotype, and function identifies the presence of an increased CD21low B-cell population. Combined with identification of the disease-causing variant, this distinguishes between healthy subjects, asymptomatic carriers, and clinically affected cases. CONCLUSION: We show that heterozygous loss-of-function variants in NFKB1 are the most common known monogenic cause of CVID, which results in a temporally progressive defect in the formation of immunoglobulin-producing B cells.


Subject(s)
B-Lymphocytes/immunology , Common Variable Immunodeficiency/genetics , NF-kappa B p50 Subunit/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Europe , Female , Humans , Infant , Infant, Newborn , Loss of Function Mutation , Male , Middle Aged , Phenotype , T-Lymphocytes/immunology , Young Adult
8.
Nat Genet ; 49(2): 262-268, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28067912

ABSTRACT

For most immune-mediated diseases, the main determinant of patient well-being is not the diagnosis itself but instead the course that the disease takes over time (prognosis). Prognosis may vary substantially between patients for reasons that are poorly understood. Familial studies support a genetic contribution to prognosis, but little evidence has been found for a proposed association between prognosis and the burden of susceptibility variants. To better characterize how genetic variation influences disease prognosis, we performed a within-cases genome-wide association study in two cohorts of patients with Crohn's disease. We identified four genome-wide significant loci, none of which showed any association with disease susceptibility. Conversely, the aggregated effect of all 170 disease susceptibility loci was not associated with disease prognosis. Together, these data suggest that the genetic contribution to prognosis in Crohn's disease is largely independent of the contribution to disease susceptibility and point to a biology of prognosis that could provide new therapeutic opportunities.


Subject(s)
Crohn Disease/genetics , Genetic Predisposition to Disease/genetics , Female , Genome/genetics , Genome-Wide Association Study/methods , Genotype , Humans , Male , Polymorphism, Single Nucleotide/genetics , Prognosis
9.
Sci Rep ; 6: 37201, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27853307

ABSTRACT

Defects in the control of Wnt signaling have emerged as a recurrent mechanism involved in cancer pathogenesis and acute myeloid leukaemia (AML), including the hematopoietic regeneration-associated WNT10B in AC133bright leukaemia cells, although the existence of a specific mechanism remains unproven. We have obtained evidences for a recurrent rearrangement, which involved the WNT10B locus (WNT10BR) within intron 1 (IVS1) and flanked at the 5' by non-human sequences whose origin remains to be elucidated; it also expressed a transcript variant (WNT10BIVS1) which was mainly detected in a cohort of patients with intermediate/unfavorable risk AML. We also identified in two separate cases, affected by AML and breast cancer respectively, a genomic transposable short form of human WNT10B (ht-WNT10B). The intronless ht-WNT10B resembles a long non-coding RNA (lncRNA), which suggests its involvement in a non-random microhomology-mediated recombination generating the rearranged WNT10BR. Furthermore, our studies supports an autocrine activation primed by the formation of WNT10B-FZD4/5 complexes in the breast cancer MCF7 cells that express the WNT10BIVS1. Chemical interference of WNT-ligands production by the porcupine inhibitor IWP-2 achieved a dose-dependent suppression of the WNT10B-FZD4/5 interactions. These results present the first evidence for a recurrent rearrangement promoted by a mobile ht-WNT10B oncogene, as a relevant mechanism for Wnt involvement in human cancer.


Subject(s)
Gene Expression Regulation, Leukemic , Gene Rearrangement , Genetic Loci , Introns , Leukemia, Myeloid, Acute , Proto-Oncogene Proteins , Wnt Proteins , Animals , Female , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Male , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Wnt Proteins/biosynthesis , Wnt Proteins/genetics , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/biosynthesis , Zebrafish Proteins/genetics
10.
Nat Neurosci ; 17(12): 1804-15, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25383901

ABSTRACT

The complexity of the human brain derives from the intricate interplay of molecular instructions during development. Here we systematically investigated gene expression changes in the prenatal human striatum and cerebral cortex during development from post-conception weeks 2 to 20. We identified tissue-specific gene coexpression networks, differentially expressed genes and a minimal set of bimodal genes, including those encoding transcription factors, that distinguished striatal from neocortical identities. Unexpected differences from mouse striatal development were discovered. We monitored 36 determinants at the protein level, revealing regional domains of expression and their refinement, during striatal development. We electrophysiologically profiled human striatal neurons differentiated in vitro and determined their refined molecular and functional properties. These results provide a resource and opportunity to gain global understanding of how transcriptional and functional processes converge to specify human striatal and neocortical neurons during development.


Subject(s)
Corpus Striatum/embryology , Corpus Striatum/physiology , Fetal Development/physiology , Gene Regulatory Networks/physiology , Action Potentials/physiology , Cell Differentiation/physiology , Cells, Cultured , HEK293 Cells , Humans , Organ Culture Techniques
11.
BMC Genomics ; 15: 649, 2014 Aug 04.
Article in English | MEDLINE | ID: mdl-25091430

ABSTRACT

BACKGROUND: Although numerous investigations have compared gene expression microarray platforms, preprocessing methods and batch correction algorithms using constructed spike-in or dilution datasets, there remains a paucity of studies examining the properties of microarray data using diverse biological samples. Most microarray experiments seek to identify subtle differences between samples with variable background noise, a scenario poorly represented by constructed datasets. Thus, microarray users lack important information regarding the complexities introduced in real-world experimental settings. The recent development of a multiplexed, digital technology for nucleic acid measurement enables counting of individual RNA molecules without amplification and, for the first time, permits such a study. RESULTS: Using a set of human leukocyte subset RNA samples, we compared previously acquired microarray expression values with RNA molecule counts determined by the nCounter Analysis System (NanoString Technologies) in selected genes. We found that gene measurements across samples correlated well between the two platforms, particularly for high-variance genes, while genes deemed unexpressed by the nCounter generally had both low expression and low variance on the microarray. Confirming previous findings from spike-in and dilution datasets, this "gold-standard" comparison demonstrated signal compression that varied dramatically by expression level and, to a lesser extent, by dataset. Most importantly, examination of three different cell types revealed that noise levels differed across tissues. CONCLUSIONS: Microarray measurements generally correlate with relative RNA molecule counts within optimal ranges but suffer from expression-dependent accuracy bias and precision that varies across datasets. We urge microarray users to consider expression-level effects in signal interpretation and to evaluate noise properties in each dataset independently.


Subject(s)
Gene Expression Profiling/methods , Oligonucleotide Array Sequence Analysis/methods , RNA/genetics , Statistics as Topic/methods , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/genetics , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/immunology , Case-Control Studies , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Leukocytes/metabolism , Organ Specificity
12.
Dev Dyn ; 243(10): 1352-61, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24801179

ABSTRACT

BACKGROUND: The transcription factor Rx1, also known as Rax, controls key properties of retinal precursors including migration behavior, proliferation, and maintenance of multipotency. However, Rx1 effector genes are largely unknown. RESULTS: To identify genes controlled by Rx1 in early retinal precursors, we compared the transcriptome of Xenopus embryos overexpressing Rx1 to that of embryos in which Rx1 was knocked-down. In particular, we selected 52 genes coherently regulated, i.e., actived in Rx1 gain of function and repressed in Rx1 loss of function experiments, or vice versa. RT-qPCR and in situ hybridization confirmed the trend of regulation predicted by microarray data for the selected genes. Most of the genes upregulated by Rx1 are coexpressed with this transcription factor, while downregulated genes are either not expressed or expressed at very low levels in the early developing retina. Putative direct Rx1 target genes, activated by GR-Rx1 in the absence of protein synthesis, include Ephrin B1 and Sh2d3c, an interactor of ephrinB1 receptor, which represent candidate novel effectors for the migration promoting activity of Rx1. CONCLUSIONS: This study identifies previously undescribed Rx1 regulated genes mainly involved in transcription regulation, cell migration/adhesion, and cell proliferation that contribute to delineate the molecular mechanisms underlying Rx1 activities.


Subject(s)
Eye Proteins/physiology , Gene Expression Regulation, Developmental , Retina/embryology , Transcriptome , Xenopus Proteins/physiology , Animals , Animals, Genetically Modified , Cell Differentiation/genetics , Embryo, Nonmammalian , Eye Proteins/genetics , Eye Proteins/metabolism , Gene Expression Profiling , Microarray Analysis , Retina/metabolism , Xenopus/embryology , Xenopus/genetics , Xenopus laevis/embryology , Xenopus laevis/genetics
13.
Stem Cells ; 31(12): 2842-7, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24038725

ABSTRACT

The molecular mechanisms underlying the acquisition of retinal precursor identity are scarcely defined. Although the homeobox gene Rx1 (also known as Rax) plays a major role in specifying retinal precursors and maintaining their multipotent state, the involved mechanisms remain to be largely deciphered. Here, following a highthroughput screen for genes regulated by Rx1, we found that this transcription factor specifies the fate of retinal progenitors by repressing genes normally activated in adjacent ectodermal territories. Unexpectedly, we also observed that Rx1, mainly through the activation of the transcriptional repressors TLE2 and Hes4, is necessary and sufficient to inhibit endomesodermal gene expression in retinal precursors of the eye field. In particular, Rx1 knockdown leads retinogenic blastomeres to adopt an endomesodermal fate, indicating a previously undescribed function for Rx1 in preventing the expression of endomesoderm determinants known to inhibit retinal fate. Altogether these data suggest that an essential requirement to establish a retinal precursor identity is the active inhibition of pathways leading to alternative fates.


Subject(s)
Eye Proteins/metabolism , Repressor Proteins/metabolism , Retina/metabolism , Transcription Factors/metabolism , Xenopus Proteins/metabolism , Animals , Cell Differentiation , Gene Expression Regulation, Developmental , Retina/cytology , Xenopus laevis
14.
J Biotechnol ; 150(1): 11-21, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20688113

ABSTRACT

We describe the use of DNA transposons as tools for carrying out functional screenings in murine embryonic stem (ES) cell-derived neural stem (NS) cells. NS cells are a new type of stem cells featuring radial glial properties, that undergoes symmetric cell division for an indefinite number of passages, expanding as a monolayer. In this model, the previously unreported Sleeping Beauty transposase M3A achieves an optimal blend of clone generation efficiency and low redundancy of integrations per clone, compared to the SB100X Sleeping Beauty variant and to the piggyBac transposon. The technology described here makes it possible to randomly trap genes in the NS cell genome and modify their expression or tag them with fluorescent markers and selectable genes, allowing recombinant cells to be isolated and expanded clonally. This approach will facilitate the identification of novel determinants of stem cell biology and neural cell fate specification in NS cells.


Subject(s)
DNA Transposable Elements/genetics , Models, Genetic , Mutagenesis, Insertional/methods , Neural Stem Cells/physiology , Transposases/genetics , Animals , Cells, Cultured , Computer Simulation , Humans , Mice , Neomycin , Transposases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL