Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Methods Mol Biol ; 2081: 161-175, 2020.
Article in English | MEDLINE | ID: mdl-31721124

ABSTRACT

In vivo bioluminescent imaging allows the detection of reporter gene expression in rodents in real time. Here we describe a novel technology whereby we can generate somatotransgenic rodents with the use of a viral vector carrying a luciferase transgene. We are able to achieve long term luciferase expression by a single injection of lentiviral or adeno-associated virus vectors to newborn mice. Further, we describe whole body bioluminescence imaging of conscious mice in a noninvasive manner, thus enforcing the 3R's (replacement, reduction, and refinement) of biomedical animal research.


Subject(s)
Gene Expression , Genes, Reporter , Luminescent Measurements/methods , Animals , Biosensing Techniques , Gene Order , Genetic Vectors/genetics , Luciferases, Firefly/genetics , Mice , Plasmids/genetics , Transfection , Transgenes
2.
Sci Rep ; 10(1): 2121, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32034258

ABSTRACT

We have previously designed a library of lentiviral vectors to generate somatic-transgenic rodents to monitor signalling pathways in diseased organs using whole-body bioluminescence imaging, in conscious, freely moving rodents. We have now expanded this technology to adeno-associated viral vectors. We first explored bio-distribution by assessing GFP expression after neonatal intravenous delivery of AAV8. We observed widespread gene expression in, central and peripheral nervous system, liver, kidney and skeletal muscle. Next, we selected a constitutive SFFV promoter and NFκB binding sequence for bioluminescence and biosensor evaluation. An intravenous injection of AAV8 containing firefly luciferase and eGFP under transcriptional control of either element resulted in strong and persistent widespread luciferase expression. A single dose of LPS-induced a 10-fold increase in luciferase expression in AAV8-NFκB mice and immunohistochemistry revealed GFP expression in cells of astrocytic and neuronal morphology. Importantly, whole-body bioluminescence persisted up to 240 days. We have validated a novel biosensor technology in an AAV system by using an NFκB response element and revealed its potential to monitor signalling pathway in a non-invasive manner in a model of LPS-induced inflammation. This technology complements existing germline-transgenic models and may be applicable to other rodent disease models.


Subject(s)
Dependovirus/genetics , Genetic Vectors/genetics , Mice, Transgenic/genetics , Animals , Biosensing Techniques/methods , Gene Expression/genetics , Green Fluorescent Proteins/genetics , Inflammation/genetics , Luciferases, Firefly/genetics , Mice , NF-kappa B/genetics , Promoter Regions, Genetic/genetics , Signal Transduction/genetics , Spleen Focus-Forming Viruses/genetics , Transcription, Genetic/genetics
3.
Stem Cell Reports ; 10(6): 1766-1781, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29681545

ABSTRACT

Human neural development begins at embryonic day 19 and marks the beginning of organogenesis. Neural stem cells in the neural tube undergo profound functional, morphological, and metabolic changes during neural specification, coordinated by a combination of exogenous and endogenous cues. The temporal cell signaling activities that mediate this process, during development and in the postnatal brain, are incompletely understood. We have applied gene expression studies and transcription factor-activated reporter lentiviruses during in vitro neural specification of human pluripotent stem cells. We show that nuclear factor κB orchestrates a multi-faceted metabolic program necessary for the maturation of neural progenitor cells during neurogenesis.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Energy Metabolism , NF-kappa B/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Autophagy , Biomarkers , Cell Cycle , Cell Differentiation/genetics , Cells, Cultured , Computational Biology/methods , Gene Expression Profiling , Gene Ontology , Humans , Immunohistochemistry , Models, Biological , Neurogenesis/genetics , Phenotype , Signal Transduction
4.
Curr Stem Cell Rep ; 3(2): 124-136, 2017.
Article in English | MEDLINE | ID: mdl-28596938

ABSTRACT

PURPOSE OF REVIEW: Alternative approaches to conventional drug-based cancer treatments have seen T cell therapies deployed more widely over the last decade. This is largely due to their ability to target and kill specific cell types based on receptor recognition. Introduction of recombinant T cell receptors (TCRs) using viral vectors and HLA-independent T cell therapies using chimeric antigen receptors (CARs) are discussed. This article reviews the tools used for genome editing, with particular emphasis on the applications of site-specific DNA nuclease mediated editing for T cell therapies. RECENT FINDINGS: Genetic engineering of T cells using TCRs and CARs with redirected antigen-targeting specificity has resulted in clinical success of several immunotherapies. In conjunction, the application of genome editing technologies has resulted in the generation of HLA-independent universal T cells for allogeneic transplantation, improved T cell sustainability through knockout of the checkpoint inhibitor, programmed cell death protein-1 (PD-1), and has shown efficacy as an antiviral therapy through direct targeting of viral genomic sequences and entry receptors. SUMMARY: The combined use of engineered antigen-targeting moieties and innovative genome editing technologies have recently shown success in a small number of clinical trials targeting HIV and hematological malignancies and are now being incorporated into existing strategies for other immunotherapies.

5.
Methods Mol Biol ; 1651: 49-64, 2017.
Article in English | MEDLINE | ID: mdl-28801899

ABSTRACT

The application of luciferase reporter genes to provide quantitative outputs for the activation of promoters is a well-established technique in molecular biology. Luciferase catalyzes an enzymatic reaction, which in the presence of the substrate luciferin produces photons of light relative to its molar concentration. The luciferase transgene can be genetically inserted at the first intron of a target gene to act as a surrogate for the gene's endogenous expression in cells and transgenic mice. Alternatively, promoter sequences can be excised and/or amplified from genomic sources or constructed de novo and cloned upstream of luciferase in an expression cassette transfected into cells. More recently, the development of synthetic promoters where the essential components of an RNA polymerase binding site and transcriptional start site are fused with various upstream regulatory sequences are being applied to drive reporter gene expression. We have developed a high-throughput cloning strategy to develop lentiviral luciferase reporters driven by transcription factor activated synthetic promoters. Lentiviruses integrate their payload cassette into the host cell genome, thereby facilitating the study of gene expression not only in the transduced cells but also within all subsequent daughter cells. In this manuscript we describe the design, vector construction, lentiviral transduction, and luciferase quantitation of transcription factor activated reporters (TFARs) in vitro and in vivo.


Subject(s)
Genes, Reporter , Luciferases, Firefly/analysis , Luminescent Agents/analysis , Luminescent Measurements/methods , Promoter Regions, Genetic , Transcriptional Activation , Animals , Cloning, Molecular , Fireflies/enzymology , Fireflies/genetics , HEK293 Cells , Humans , Lentivirus/genetics , Luciferases, Firefly/genetics , Luminescent Agents/metabolism , Mice , Transcription Factors/metabolism , Transduction, Genetic/methods , Transgenes
6.
Sci Rep ; 7: 41874, 2017 02 03.
Article in English | MEDLINE | ID: mdl-28157201

ABSTRACT

Molecular mechanisms regulating liver repair following cholestatic injury remain largely unknown. We have combined a mouse model of acute cholestatic liver injury, partial bile duct ligation (pBDL), with a novel longitudinal bioimaging methodology to quantify transcription factor activity during hepatic injury and repair. We administered lentiviral transcription factor activated luciferase/eGFP reporter (TFAR) cassettes to neonatal mice enabling longitudinal TFAR profiling by continued bioimaging throughout the lives of the animals and following pBDL in adulthood. Neonatal intravascular injection of VSV-G pseudotyped lentivirus resulted in almost exclusive transduction of hepatocytes allowing analysis of hepatocyte-specific transcription factor activity. We recorded acute but transient responses with NF-κB and Smad2/3 TFAR whilst our Notch reporter was repressed over the 40 days of evaluation post-pBDL. The bipotent hepatic progenitor cell line, HepaRG, can be directed to differentiate into hepatocytes and biliary epithelia. We found that forced expression of the Notch inhibitor NUMB in HepaRG resulted in enhanced hepatocyte differentiation and proliferation whereas over-expressing the Notch agonist JAG1 resulted in biliary epithelial differentiation. In conclusion, our data demonstrates that hepatocytes rapidly upregulate NF-κB and Smad2/3 activity, whilst repressing Notch signalling. This transcriptional response to cholestatic liver injury likely promotes partial de-differentiation to allow pro-regenerative proliferation of hepatocytes.


Subject(s)
Cholestasis/metabolism , Hepatocytes/metabolism , Liver Diseases/metabolism , Signal Transduction , 3T3 Cells , Animals , Cell Differentiation , Cell Proliferation , Cholestasis/complications , Cholestasis/diagnostic imaging , Epithelial Cells/cytology , Epithelial Cells/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Hepatocytes/cytology , Hepatocytes/physiology , Humans , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Lentivirus/genetics , Liver Diseases/diagnostic imaging , Liver Diseases/etiology , Luciferases/genetics , Luciferases/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism
7.
Sci Rep ; 7(1): 6374, 2017 07 25.
Article in English | MEDLINE | ID: mdl-28743959

ABSTRACT

Luciferase bioimaging in living animals is increasingly being applied in many fields of biomedical research. Rodent imaging usually involves anaesthetising the animal during data capture, however, the biological consequences of anaesthesia have been largely overlooked. We have evaluated luciferase bioimaging in conscious, unrestrained mice after neonatal intracranial or intravascular administration of lentiviral, luciferase reporter cassettes (biosensors); we present real-time analyses from the first day of life to adulthood. Anaesthetics have been shown to exert both neurotoxic and neuroprotective effects during development and in models of brain injury. Mice subjected to bioimaging after neonatal intracranial or intravascular administration of biosensors, targeting the brain and liver retrospectively showed no significant difference in luciferase expression when conscious or unconscious throughout development. We applied conscious bioimaging to the assessment of NFκB and STAT3 transcription factor activated reporters during the earliest stages of development in living, unrestrained pups. Our data showed unique longitudinal activities for NFκB and STAT3 in the brain of conscious mice. Conscious bioimaging was applied to a neonatal mouse model of cerebral palsy (Hypoxic-Ischaemic Encephalopathy). Imaging of NFκB reporter before and after surgery showed a significant increase in luciferase expression, coinciding with secondary energy failure, in lesioned mice compared to controls.


Subject(s)
Brain/metabolism , Cerebral Palsy/metabolism , Liver/metabolism , Luciferases/metabolism , Molecular Imaging/methods , Animals , Animals, Newborn , Biosensing Techniques/methods , Cerebral Palsy/surgery , Consciousness , Disease Models, Animal , Genetic Vectors/administration & dosage , Injections, Intra-Arterial , Lentivirus/genetics , Luciferases/genetics , Mice , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/metabolism , Recombinant Proteins/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
8.
Cell Rep ; 14(8): 1883-91, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26904936

ABSTRACT

The potential of induced pluripotent stem cells (iPSCs) in disease modeling and regenerative medicine is vast, but current methodologies remain inefficient. Understanding the cellular mechanisms underlying iPSC reprogramming, such as the metabolic shift from oxidative to glycolytic energy production, is key to improving its efficiency. We have developed a lentiviral reporter system to assay longitudinal changes in cell signaling and transcription factor activity in living cells throughout iPSC reprogramming of human dermal fibroblasts. We reveal early NF-κB, AP-1, and NRF2 transcription factor activation prior to a temporal peak in hypoxia inducible factor α (HIFα) activity. Mechanistically, we show that an early burst in oxidative phosphorylation and elevated reactive oxygen species generation mediates increased NRF2 activity, which in turn initiates the HIFα-mediated glycolytic shift and may modulate glucose redistribution to the pentose phosphate pathway. Critically, inhibition of NRF2 by KEAP1 overexpression compromises metabolic reprogramming and results in reduced efficiency of iPSC colony formation.


Subject(s)
Cellular Reprogramming , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/metabolism , Kelch-Like ECH-Associated Protein 1/genetics , NF-E2-Related Factor 2/genetics , Dermis/cytology , Dermis/metabolism , Fibroblasts/cytology , Gene Expression Regulation , Genes, Reporter , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glycolysis/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Induced Pluripotent Stem Cells/cytology , Kelch-Like ECH-Associated Protein 1/metabolism , Lentivirus/genetics , Lentivirus/metabolism , Luciferases/genetics , Luciferases/metabolism , NF-E2-Related Factor 2/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Oxidative Phosphorylation , Pentose Phosphate Pathway/genetics , Reactive Oxygen Species/metabolism , Signal Transduction , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transduction, Genetic
9.
Sci Rep ; 5: 11842, 2015 Jul 03.
Article in English | MEDLINE | ID: mdl-26138224

ABSTRACT

The application of transcription factor activated luciferase reporter cassettes in vitro is widespread but potential for in vivo application has not yet been realized. Bioluminescence imaging enables non-invasive tracking of gene expression in transfected tissues of living rodents. However the mature immune response limits luciferase expression when delivered in adulthood. We present a novel approach of tissue-targeted delivery of transcription factor activated luciferase reporter lentiviruses to neonatal rodents as an alternative to the existing technology of generating germline transgenic light producing rodents. At this age, neonates acquire immune tolerance to the conditionally responsive luciferase reporter. This simple and transferrable procedure permits surrogate quantitation of transcription factor activity over the lifetime of the animal. We show principal efficacy by temporally quantifying NFκB activity in the brain, liver and lungs of somatotransgenic reporter mice subjected to lipopolysaccharide (LPS)-induced inflammation. This response is ablated in Tlr4(-/-) mice or when co-administered with the anti-inflammatory glucocorticoid analogue dexamethasone. Furthermore, we show the malleability of this technology by quantifying NFκB-mediated luciferase expression in outbred rats. Finally, we use somatotransgenic bioimaging to longitudinally quantify LPS- and ActivinA-induced upregulation of liver specific glucocorticoid receptor and Smad2/3 reporter constructs in somatotransgenic mice, respectively.


Subject(s)
Luciferases, Firefly/genetics , Transcription Factors/physiology , Transcriptional Activation/immunology , Animals , Genes, Reporter , Genetic Vectors , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , Lentivirus/genetics , Lipopolysaccharides/pharmacology , Luciferases, Firefly/biosynthesis , Mice , NIH 3T3 Cells , Organ Specificity , Rats, Sprague-Dawley , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics
10.
Methods Mol Biol ; 891: 273-90, 2012.
Article in English | MEDLINE | ID: mdl-22648777

ABSTRACT

Surrogate genetically encoded markers have been utilized in order to analyze gene transfer efficacy, location, and persistence. These marker genes have greatly accelerated the development of gene transfer vectors for the ultimate application of gene therapy using therapeutic genes. They have also been used in many other applications, such as gene marking in order to study developmental cell lineages, to track cell migration, and to study tumor growth and metastasis. This chapter aims to describe the analysis of several commonly used marker genes: green fluorescent protein (GFP), ß-galactosidase, firefly luciferase, human factor IX, and alkaline phosphatase. The merits and disadvantages of each are briefly discussed. In addition a few short examples are provided for continual and endpoint analysis in different disease models including hemophilia, cystic fibrosis, ornithine transcarbamylase deficiency and Gaucher disease.


Subject(s)
Biomarkers/metabolism , Genetic Therapy/methods , Prenatal Care/methods , Alkaline Phosphatase/metabolism , Animals , DNA/isolation & purification , Enzyme Assays , Enzyme-Linked Immunosorbent Assay , Factor IX/metabolism , Genetic Vectors/genetics , Green Fluorescent Proteins/metabolism , Humans , Immunohistochemistry , Luciferases, Firefly/metabolism , Mice , Polymerase Chain Reaction , Staining and Labeling , Tissue Extracts , Whole Body Imaging , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL