Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Blood ; 2024 May 28.
Article in English | MEDLINE | ID: mdl-38805639

ABSTRACT

Loss of long-term hematopoietic stem cell (LT-HSC) function ex vivo hampers the success of clinical protocols reliant on culture. However, the kinetics and mechanisms by which this occurs remain incompletely characterized. Here, through time-resolved scRNA-Seq, matched in vivo functional analysis and the use of a reversible in vitro system of early G1 arrest, we define the sequence of transcriptional and functional events occurring during the first ex vivo division of human LT-HSCs. We demonstrate that the sharpest loss of LT-HSC repopulation capacity happens early on, between 6 and 24 hours of culture, before LT-HSCs commit to cell cycle progression. During this time window, LT-HSCs adapt to the culture environment, limiting global variability in gene expression and transiently upregulating gene networks involved in signaling and stress responses. From 24 hours, LT-HSC progression past early G1 contributes to the establishment of differentiation programmes in culture. However, contrary to current assumptions, we demonstrate that loss of HSC function ex vivo is independent of cell cycle progression. Finally, we show that targeting LT-HSC adaptation to culture by inhibiting early activation of JAK/STAT signaling improves HSC long-term repopulating function ex vivo. Collectively, our study demonstrates that controlling early LT-HSC adaptation to ex vivo culture, for example via JAK inhibition, is of critical importance to improve HSC gene therapy and expansion protocols.

2.
Mol Ther Methods Clin Dev ; 19: 47-57, 2020 Dec 11.
Article in English | MEDLINE | ID: mdl-32995359

ABSTRACT

Stable suspension producer cell lines for the production of vesicular stomatitis virus envelope glycoprotein (VSVg)-pseudotyped lentiviral vectors represent an attractive alternative to current widely used production methods based on transient transfection of adherent 293T cells with multiple plasmids. We report here a method to rapidly generate such producer cell lines from 293T cells by stable transfection of a single DNA construct encoding all lentiviral vector components. The resulting suspension cell lines yield titers as high as can be achieved with transient transfection, can be readily scaled up in single-use stirred-tank bioreactors, and are genetically and functionally stable in extended cell culture. By removing the requirement for efficient transient transfection during upstream processing of lentiviral vectors and switching to an inherently scalable suspension cell culture format, we believe that this approach will result in significantly higher batch yields than are possible with current manufacturing processes and enable better patient access to medicines based on lentiviral vectors.

3.
Mol Ther ; 27(10): 1706-1717, 2019 10 02.
Article in English | MEDLINE | ID: mdl-31526597

ABSTRACT

The field of cell and gene therapy (GT) is expanding rapidly and there is undoubtedly a wave of enthusiasm and anticipation for what these treatments could achieve next. Here we assessed the worldwide landscape of GT assets currently in early clinical development (clinical trial phase 1/2 or about to enter clinical trial). We included all gene therapies, i.e., strategies that modify an individual's protein make-up by introducing exogenous nucleic acid or nucleic acid modifiers, regardless of delivery. Unmodified cell therapies, oncology therapies (reviewed elsewhere), and vaccine programs (distinct therapeutic strategy) were not included. Using a December 31, 2018 cutoff date, we identified 336 gene therapies being developed for 138 different indications covering 165 genetic targets. In all, we found that the early clinical GT landscape comprises a very disparate group of drug candidates in terms of indications, organizations, and delivery methods. We also highlight interesting trends, revealing the evolution of the field toward in vivo therapies and adeno-associated virus vector-based delivery systems. It will be interesting to witness what proportion of this current list effectively translates into new medicines.


Subject(s)
Drug Delivery Systems/classification , Genetic Therapy/methods , Clinical Trials as Topic , Genetic Vectors/administration & dosage , Humans , Molecular Targeted Therapy
4.
Int J Mol Sci ; 20(18)2019 Sep 05.
Article in English | MEDLINE | ID: mdl-31491876

ABSTRACT

Fabry disease (FD) is caused by mutations in the GLA gene that encodes lysosomal α-galactosidase-A (α-gal-A). A number of pathogenic mechanisms have been proposed and these include loss of mitochondrial respiratory chain activity. For FD, gene therapy is beginning to be applied as a treatment. In view of the loss of mitochondrial function reported in FD, we have considered here the impact of loss of mitochondrial respiratory chain activity on the ability of a GLA lentiviral vector to increase cellular α-gal-A activity and participate in cross correction. Jurkat cells were used in this study and were exposed to increasing viral copies. Intracellular and extracellular enzyme activities were then determined; this in the presence or absence of the mitochondrial complex I inhibitor, rotenone. The ability of cells to take up released enzyme was also evaluated. Increasing transgene copies was associated with increasing intracellular α-gal-A activity but this was associated with an increase in Km. Release of enzyme and cellular uptake was also demonstrated. However, in the presence of rotenone, enzyme release was inhibited by 37%. Excessive enzyme generation may result in a protein with inferior kinetic properties and a background of compromised mitochondrial function may impair the cross correction process.


Subject(s)
Electron Transport Complex I/genetics , Mitochondria/genetics , Mitochondria/metabolism , alpha-Galactosidase/biosynthesis , Cell Line , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/metabolism , Fabry Disease/genetics , Fabry Disease/metabolism , Gene Dosage , Gene Expression , Humans , Jurkat Cells , Lysosomes/metabolism , Mitochondria/drug effects , Transduction, Genetic , Transgenes , alpha-Galactosidase/genetics
5.
Mol Ther Nucleic Acids ; 12: 626-634, 2018 Sep 07.
Article in English | MEDLINE | ID: mdl-30081233

ABSTRACT

Viral vectors are rapidly being developed for a range of applications in research and gene therapy. Prototype foamy virus (PFV) vectors have been described for gene therapy, although their use has mainly been restricted to ex vivo stem cell modification. Here we report direct in vivo transgene delivery with PFV vectors carrying reporter gene constructs. In our investigations, systemic PFV vector delivery to neonatal mice gave transgene expression in the heart, xiphisternum, liver, pancreas, and gut, whereas intracranial administration produced brain expression until animals were euthanized 49 days post-transduction. Immunostaining and confocal microscopy analysis of injected brains showed that transgene expression was highly localized to hippocampal architecture despite vector delivery being administered to the lateral ventricle. This was compared with intracranial biodistribution of lentiviral vectors and adeno-associated virus vectors, which gave a broad, non-specific spread through the neonatal mouse brain without regional localization, even when administered at lower copy numbers. Our work demonstrates that PFV can be used for neonatal gene delivery with an intracranial expression profile that localizes to hippocampal neurons, potentially because of the mitotic status of the targeted cells, which could be of use for research applications and gene therapy of neurological disorders.

6.
Exp Hematol ; 57: 21-29, 2018 01.
Article in English | MEDLINE | ID: mdl-28911908

ABSTRACT

Integration-deficient lentiviruses (IdLVs) deliver genes effectively to tissues but are lost rapidly from dividing cells. This property can be harnessed to express transgenes transiently to manipulate cell biology. Here, we demonstrate the utility of short-term gene expression to improve functional potency of hematopoietic stem and progenitor cells (HSPCs) during transplantation by delivering HOXB4 and Angptl3 using IdLVs to enhance the engraftment of HSPCs. Constitutive overexpression of either of these genes is likely to be undesirable, but the transient nature of IdLVs reduces this risk and those associated with unsolicited gene expression in daughter cells. Transient expression led to increased multilineage hematopoietic engraftment in in vivo competitive repopulation assays without the side effects reported in constitutive overexpression models. Adult stem cell fate has not been programmed previously using IdLVs, but we demonstrate that these transient gene expression tools can produce clinically relevant alterations or be applied to investigate basic biology.


Subject(s)
Genetic Vectors/genetics , Hematopoietic Stem Cells/physiology , Lentivirus/genetics , Transduction, Genetic , Angiopoietin-Like Protein 3 , Angiopoietin-like Proteins/biosynthesis , Angiopoietin-like Proteins/genetics , Animals , Cell Lineage , Gene Expression Regulation , Genes, Reporter , Graft Survival , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , K562 Cells , Mice , Mice, Inbred C57BL , Radiation Chimera , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transgenes
7.
Sci Rep ; 7(1): 12475, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28963568

ABSTRACT

The generation of T cells from pluripotent stem cells (PSCs) is attractive for investigating T cell development and validating genome editing strategies in vitro. X-linked severe combined immunodeficiency (X-SCID) is an immune disorder caused by mutations in the IL2RG gene and characterised by the absence of T and NK cells in patients. IL2RG encodes the common gamma chain, which is part of several interleukin receptors, including IL-2 and IL-7 receptors. To model X-SCID in vitro, we generated a mouse embryonic stem cell (ESC) line in which a disease-causing human IL2RG gene variant replaces the endogenous Il2rg locus. We developed a stage-specific T cell differentiation protocol to validate genetic correction of the common G691A mutation with transcription activator-like effector nucleases. While all ESC clones could be differentiated to hematopoietic precursor cells, stage-specific analysis of T cell maturation confirmed early arrest of T cell differentiation at the T cell progenitor stage in X-SCID cells. In contrast, genetically corrected ESCs differentiated to CD4 + or CD8 + single-positive T cells, confirming correction of the cellular X-SCID phenotype. This study emphasises the value of PSCs for disease modelling and underlines the significance of in vitro models as tools to validate genome editing strategies before clinical application.


Subject(s)
Gene Editing/methods , Hematopoietic Stem Cells/immunology , Interleukin Receptor Common gamma Subunit/genetics , Mouse Embryonic Stem Cells/immunology , X-Linked Combined Immunodeficiency Diseases/genetics , X-Linked Combined Immunodeficiency Diseases/therapy , Amino Acid Substitution , Animals , Cell Differentiation , Disease Models, Animal , Gene Expression , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/pathology , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Interleukin-2/pharmacology , Interleukin-7/genetics , Interleukin-7/immunology , Interleukin-7/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Mice , Mice, SCID , Mice, Transgenic , Molecular Targeted Therapy , Mouse Embryonic Stem Cells/drug effects , Mouse Embryonic Stem Cells/pathology , Mutation , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Transcription Activator-Like Effector Nucleases/genetics , Transcription Activator-Like Effector Nucleases/immunology , Transgenes , X-Linked Combined Immunodeficiency Diseases/immunology , X-Linked Combined Immunodeficiency Diseases/pathology
9.
Mol Ther ; 25(8): 1790-1804, 2017 08 02.
Article in English | MEDLINE | ID: mdl-28550974

ABSTRACT

Lentiviral vector genomic RNA requires sequences that partially overlap wild-type HIV-1 gag and env genes for packaging into vector particles. These HIV-1 packaging sequences constitute 19.6% of the wild-type HIV-1 genome and contain functional cis elements that potentially compromise clinical safety. Here, we describe the development of a novel lentiviral vector (LTR1) with a unique genomic structure designed to prevent transfer of HIV-1 packaging sequences to patient cells, thus reducing the total HIV-1 content to just 4.8% of the wild-type genome. This has been achieved by reconfiguring the vector to mediate reverse-transcription with a single strand transfer, instead of the usual two, and in which HIV-1 packaging sequences are not copied. We show that LTR1 vectors offer improved safety in their resistance to remobilization in HIV-1 particles and reduced frequency of splicing into human genes. Following intravenous luciferase vector administration to neonatal mice, LTR1 sustained a higher level of liver transgene expression than an equivalent dose of a standard lentivirus. LTR1 vectors produce reverse-transcription products earlier and start to express transgenes significantly quicker than standard lentiviruses after transduction. Finally, we show that LTR1 is an effective lentiviral gene therapy vector as demonstrated by correction of a mouse hemophilia B model.


Subject(s)
Gene Transfer Techniques , Genetic Vectors/genetics , HIV-1/genetics , RNA, Viral , Regulatory Sequences, Ribonucleic Acid , Transduction, Genetic , Animals , Cell Line , Disease Models, Animal , Factor IX/genetics , Gene Expression , Gene Order , Genes, Reporter , Genetic Therapy , Genome, Viral , HIV Long Terminal Repeat , Hemophilia B/blood , Hemophilia B/genetics , Hemophilia B/therapy , Humans , Mice , Proviruses/genetics , Recombination, Genetic , Transgenes , Virus Replication/genetics
10.
Sci Rep ; 7(1): 79, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28250438

ABSTRACT

Duchenne Muscular Dystrophy (DMD) is caused by a lack of dystrophin expression in patient muscle fibres. Current DMD gene therapy strategies rely on the expression of internally deleted forms of dystrophin, missing important functional domains. Viral gene transfer of full-length dystrophin could restore wild-type functionality, although this approach is restricted by the limited capacity of recombinant viral vectors. Lentiviral vectors can package larger transgenes than adeno-associated viruses, yet lentiviral vectors remain largely unexplored for full-length dystrophin delivery. In our work, we have demonstrated that lentiviral vectors can package and deliver inserts of a similar size to dystrophin. We report a novel approach for delivering large transgenes in lentiviruses, in which we demonstrate proof-of-concept for a 'template-switching' lentiviral vector that harnesses recombination events during reverse-transcription. During this work, we discovered that a standard, unmodified lentiviral vector was efficient in delivering full-length dystrophin to target cells, within a total genomic load of more than 15,000 base pairs. We have demonstrated gene therapy with this vector by restoring dystrophin expression in DMD myoblasts, where dystrophin was expressed at the sarcolemma of myotubes after myogenic differentiation. Ultimately, our work demonstrates proof-of-concept that lentiviruses can be used for permanent full-length dystrophin gene therapy, which presents a significant advancement in developing an effective treatment for DMD.


Subject(s)
Dystrophin/genetics , Dystrophin/metabolism , Lentivirus/genetics , Muscular Dystrophy, Duchenne/genetics , Myoblasts, Skeletal/metabolism , Cells, Cultured , DNA Packaging , Genetic Therapy , Genetic Vectors/genetics , HEK293 Cells , HeLa Cells , Humans , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/therapy , Myoblasts, Skeletal/pathology , Recombination, Genetic , Transduction, Genetic
11.
Sci Rep ; 7: 44775, 2017 03 17.
Article in English | MEDLINE | ID: mdl-28303972

ABSTRACT

Duchenne Muscular Dystrophy (DMD) is caused by a lack of dystrophin expression in patient muscle fibres. Current DMD gene therapy strategies rely on the expression of internally deleted forms of dystrophin, missing important functional domains. Viral gene transfer of full-length dystrophin could restore wild-type functionality, although this approach is restricted by the limited capacity of recombinant viral vectors. Lentiviral vectors can package larger transgenes than adeno-associated viruses, yet lentiviral vectors remain largely unexplored for full-length dystrophin delivery. In our work, we have demonstrated that lentiviral vectors can package and deliver inserts of a similar size to dystrophin. We report a novel approach for delivering large transgenes in lentiviruses, in which we demonstrate proof-of-concept for a 'template-switching' lentiviral vector that harnesses recombination events during reverse-transcription. During this work, we discovered that a standard, unmodified lentiviral vector was efficient in delivering full-length dystrophin to target cells, within a total genomic load of more than 15,000 base pairs. We have demonstrated gene therapy with this vector by restoring dystrophin expression in DMD myoblasts, where dystrophin was expressed at the sarcolemma of myotubes after myogenic differentiation. Ultimately, our work demonstrates proof-of-concept that lentiviruses can be used for permanent full-length dystrophin gene therapy, which presents a significant advancement in developing an effective treatment for DMD.


Subject(s)
Dystrophin/genetics , Dystrophin/therapeutic use , Genetic Therapy , Genetic Vectors/metabolism , Lentivirus/genetics , Cell Line , Child, Preschool , Humans , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Myoblasts/metabolism , Myoblasts/pathology , Templates, Genetic , Transduction, Genetic , Transgenes
12.
Am J Physiol Lung Cell Mol Physiol ; 312(2): L258-L267, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27979861

ABSTRACT

Air-liquid interface (ALI) culture of primary airway epithelial cells enables mucociliary differentiation providing an in vitro model of the human airway, but their proliferative potential is limited. To extend proliferation, these cells were previously transduced with viral oncogenes or mouse Bmi-1 + hTERT, but the resultant cell lines did not undergo mucociliary differentiation. We hypothesized that use of human BMI-1 alone would increase the proliferative potential of bronchial epithelial cells while retaining their mucociliary differentiation potential. Cystic fibrosis (CF) and non-CF bronchial epithelial cells were transduced by lentivirus with BMI-1 and then their morphology, replication kinetics, and karyotype were assessed. When differentiated at ALI, mucin production, ciliary function, and transepithelial electrophysiology were measured. Finally, shRNA knockdown of DNAH5 in BMI-1 cells was used to model primary ciliary dyskinesia (PCD). BMI-1-transduced basal cells showed normal cell morphology, karyotype, and doubling times despite extensive passaging. The cell lines underwent mucociliary differentiation when cultured at ALI with abundant ciliation and production of the gel-forming mucins MUC5AC and MUC5B evident. Cilia displayed a normal beat frequency and 9+2 ultrastructure. Electrophysiological characteristics of BMI-1-transduced cells were similar to those of untransduced cells. shRNA knockdown of DNAH5 in BMI-1 cells produced immotile cilia and absence of DNAH5 in the ciliary axoneme as seen in cells from patients with PCD. BMI-1 delayed senescence in bronchial epithelial cells, increasing their proliferative potential but maintaining mucociliary differentiation at ALI. We have shown these cells are amenable to genetic manipulation and can be used to produce novel disease models for research and dissemination.


Subject(s)
Bronchi/cytology , Cell Differentiation , Cilia/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Mucus/metabolism , Polycomb Repressive Complex 1/metabolism , Animals , Axonemal Dyneins/metabolism , Cell Proliferation , Cell Shape , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Dyneins/metabolism , Electric Impedance , Electrophysiological Phenomena , Gene Knockdown Techniques , HEK293 Cells , Humans , Kartagener Syndrome/metabolism , Kartagener Syndrome/pathology , Kartagener Syndrome/physiopathology , Karyotyping , Mice , Microtubules/metabolism , Models, Biological , Phenotype , Transduction, Genetic
13.
Sci Rep ; 6: 23125, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26975732

ABSTRACT

Gene therapy for cystic fibrosis using non-viral, plasmid-based formulations has been the subject of intensive research for over two decades but a clinically viable product has yet to materialise in large part due to inefficient transgene expression. Minicircle DNA give enhanced and more persistent transgene expression compared to plasmid DNA in a number of organ systems but has not been assessed in the lung. In this study we compared minicircle DNA with plasmid DNA in transfections of airway epithelial cells. In vitro, luciferase gene expression from minicircles was 5-10-fold higher than with plasmid DNA. In eGFP transfections in vitro both the mean fluorescence intensity and percentage of cells transfected was 2-4-fold higher with minicircle DNA. Administration of equimolar amounts of DNA to mouse lungs resulted in a reduced inflammatory response and more persistent transgene expression, with luciferase activity persisting for 2 weeks from minicircle DNA compared to plasmid formulations. Transfection of equal mass amounts of DNA in mouse lungs resulted in a 6-fold increase in transgene expression in addition to more persistent transgene expression. Our findings have clear implications for gene therapy of airway disorders where plasmid DNA transfections have so far proven inefficient in clinical trials.


Subject(s)
Axonemal Dyneins/genetics , DNA, Circular/genetics , Gene Transfer Techniques , Green Fluorescent Proteins/genetics , Luciferases, Firefly/genetics , Lung/metabolism , Animals , Axonemal Dyneins/metabolism , Cell Line , Cytokines/metabolism , Epithelial Cells/metabolism , Female , Gene Expression , Genetic Vectors , Green Fluorescent Proteins/metabolism , Humans , Luciferases, Firefly/metabolism , Mice , Plasmids , Transfection , Transgenes
14.
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
15.
FASEB J ; 29(9): 3876-88, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26062602

ABSTRACT

Several acute monogenic diseases affect multiple body systems, causing death in childhood. The development of novel therapies for such conditions is challenging. However, improvements in gene delivery technology mean that gene therapy has the potential to treat such disorders. We evaluated the ability of the AAV9 vector to mediate systemic gene delivery after intravenous administration to perinatal mice and late-gestation nonhuman primates (NHPs). Titer-matched single-stranded (ss) and self-complementary (sc) AAV9 carrying the green fluorescent protein (GFP) reporter gene were intravenously administered to fetal and neonatal mice, with noninjected age-matched mice used as the control. Extensive GFP expression was observed in organs throughout the body, with the epithelial and muscle cells being particularly well transduced. ssAAV9 carrying the WPRE sequence mediated significantly more gene expression than its sc counterpart, which lacked the woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) sequence. To examine a realistic scale-up to larger models or potentially patients for such an approach, AAV9 was intravenously administered to late-gestation NHPs by using a clinically relevant protocol. Widespread systemic gene expression was measured throughout the body, with cellular tropisms similar to those observed in the mouse studies and no observable adverse events. This study confirms that AAV9 can safely mediate systemic gene delivery in small and large animal models and supports its potential use in clinical systemic gene therapy protocols.


Subject(s)
Dependovirus , Fetus , Genetic Vectors , Green Fluorescent Proteins , Transduction, Genetic/methods , Viral Tropism , Animals , Female , Fetus/cytology , Fetus/embryology , Fetus/metabolism , Genetic Vectors/genetics , Genetic Vectors/metabolism , Genetic Vectors/pharmacology , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Haplorhini , Mice , Pregnancy
16.
J Immunol Res ; 2015: 397879, 2015.
Article in English | MEDLINE | ID: mdl-25759840

ABSTRACT

Following fetal or neonatal gene transfer in mice and other species immune tolerance of the transgenic protein is frequently observed; however the underlying mechanisms remain largely undefined. In this study fetal and neonatal BALB/c mice received adenovirus vector to deliver human factor IX (hFIX) cDNA. The long-term tolerance of hFIX was robust in the face of immune challenge with hFIX protein and adjuvant but was eliminated by simultaneous administration of anti-CD25+ antibody. Naive irradiated BALB/c mice which had received lymphocytes from donors immunised with hFIX developed anti-hFIX antibodies upon immune challenge. Cotransplantation with CD4+CD25+ cells isolated from neonatally tolerized donors decreased the antibody response. In contrast, cotransplantation with CD4+CD25- cells isolated from the same donors increased the antibody response. These data provide evidence that immune tolerance following perinatal gene transfer is maintained by a CD4+CD25+ regulatory population.


Subject(s)
Adenoviridae/genetics , Factor IX/genetics , Factor IX/immunology , Gene Transfer Techniques , Genetic Vectors/genetics , Immune Tolerance , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Antibodies/blood , Antibodies/immunology , CD4 Antigens/metabolism , Factor IX/metabolism , Gene Expression , Genetic Vectors/administration & dosage , Humans , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Depletion , Mice , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/metabolism , Tissue Distribution
17.
Nat Commun ; 6: 6094, 2015 Jan 23.
Article in English | MEDLINE | ID: mdl-25615415

ABSTRACT

T-cell acute lymphoblastic leukaemias (T-ALL) are aggressive malignant proliferations characterized by high relapse rates and great genetic heterogeneity. TAL1 is amongst the most frequently deregulated oncogenes. Yet, over half of the TAL1(+) cases lack TAL1 lesions, suggesting unrecognized (epi)genetic deregulation mechanisms. Here we show that TAL1 is normally silenced in the T-cell lineage, and that the polycomb H3K27me3-repressive mark is focally diminished in TAL1(+) T-ALLs. Sequencing reveals that >20% of monoallelic TAL1(+) patients without previously known alterations display microinsertions or RAG1/2-mediated episomal reintegration in a single site 5' to TAL1. Using 'allelic-ChIP' and CrispR assays, we demonstrate that such insertions induce a selective switch from H3K27me3 to H3K27ac at the inserted but not the germline allele. We also show that, despite a considerable mechanistic diversity, the mode of oncogenic TAL1 activation, rather than expression levels, impact on clinical outcome. Altogether, these studies establish site-specific epigenetic desilencing as a mechanism of oncogenic activation.


Subject(s)
Alleles , Gene Expression Regulation, Leukemic , Polycomb-Group Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Acetylation , Adult , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chromatin Immunoprecipitation , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Genetic Loci , Histones/metabolism , Homeodomain Proteins/metabolism , Humans , Jurkat Cells , Methylation , Molecular Sequence Data , Mutagenesis, Insertional , Nuclear Proteins/metabolism , Plasmids/genetics , Polycomb-Group Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Survival Analysis , T-Cell Acute Lymphocytic Leukemia Protein 1 , Treatment Outcome
18.
Methods Mol Biol ; 891: 85-107, 2012.
Article in English | MEDLINE | ID: mdl-22648769

ABSTRACT

Vectors derived from the Retroviridae family have several attributes required for successful gene delivery. Retroviral vectors have an adequate payload size for the coding regions of most genes; they are safe to handle and simple to produce. These vectors can be manipulated to target different cell types with low immunogenicity and can permanently insert genetic information into the host cells' genome. Retroviral vectors have been used in gene therapy clinical trials and successfully applied experimentally in vitro, in vivo, and in utero.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/biosynthesis , Genetic Vectors/genetics , Prenatal Care/methods , Retroviridae/genetics , Enzyme-Linked Immunosorbent Assay , Humans , Polymerase Chain Reaction , Titrimetry
19.
Mol Ther ; 20(8): 1610-21, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22547151

ABSTRACT

Mucopolysaccharidosis IIIA (MPS IIIA or Sanfilippo disease) is a neurodegenerative disorder caused by a deficiency in the lysosomal enzyme sulfamidase (SGSH), catabolizing heparan sulfate (HS). Affected children present with severe behavioral abnormalities, sleep disturbances, and progressive neurodegeneration, leading to death in their second decade. MPS I, a similar neurodegenerative disease accumulating HS, is treated successfully with hematopoietic stem cell transplantation (HSCT) but this treatment is ineffectual for MPS IIIA. We compared HSCT in MPS IIIA mice using wild-type donor cells transduced ex vivo with lentiviral vector-expressing SGSH (LV-WT-HSCT) versus wild-type donor cell transplant (WT-HSCT) or lentiviral-SGSH transduced MPS IIIA cells (LV-IIIA-HSCT). LV-WT-HSCT results in 10% of normal brain enzyme activity, near normalization of brain HS and GM2 gangliosides, significant improvements in neuroinflammation and behavioral correction. Both WT-HSCT and LV-IIIA-HSCT mediated improvements in GM2 gangliosides and neuroinflammation but were less effective at reducing HS or in ameliorating abnormal HS sulfation and had no significant effect on behavior. This suggests that HS may have a more significant role in neuropathology than neuroinflammation or GM2 gangliosides. These data provide compelling evidence for the efficacy of gene therapy in conjunction with WT-HSCT for neurological correction of MPS IIIA where conventional transplant is ineffectual.


Subject(s)
Genetic Therapy/methods , Hematopoietic Stem Cells/physiology , Mucopolysaccharidoses/pathology , Mucopolysaccharidoses/therapy , Animals , Female , Flow Cytometry , Hematopoietic Stem Cells/cytology , Immunohistochemistry , Mice
20.
Stem Cells ; 30(6): 1134-43, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22367629

ABSTRACT

Human mesenchymal stem cells (hMSCs) have been shown to have potential in regenerative approaches in bone and blood. Most protocols rely on their in vitro expansion prior to clinical use. However, several groups including our own have shown that hMSCs lose proliferation and differentiation ability with serial passage in culture, limiting their clinical applications. Cellular prion protein (PrP) has been shown to enhance proliferation and promote self-renewal of hematopoietic, mammary gland, and neural stem cells. Here we show, for the first time, that expression of PrP decreased in hMSC following ex vivo expansion. When PrP expression was knocked down, hMSC showed significant reduction in proliferation and differentiation. In contrast, hMSC expanded in the presence of small molecule 3/689, a modulator of PrP expression, showed retention of PrP expression with ex vivo expansion and extended lifespan up to 10 population doublings. Moreover, cultures produced a 300-fold increase in the number of cells generated. These cells showed a 10-fold increase in engraftment levels in bone marrow 5 weeks post-transplant. hMSC treated with 3/689 showed enhanced protection from DNA damage and enhanced cell cycle progression, in line with data obtained by gene expression profiling. Moreover, upregulation of superoxide dismutase-2 (SOD2) was also observed in hMSC expanded in the presence of 3/689. The increase in SOD2 was dependent on PrP expression and suggests increased scavenging of reactive oxygen species as mechanism of action. These data point to PrP as a good target for chemical intervention in stem cell regenerative medicine.


Subject(s)
Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Prions/biosynthesis , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/physiology , Cell Growth Processes/physiology , Cells, Cultured , Gene Knockdown Techniques , HEK293 Cells , Humans , Lentivirus/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred NOD , Mice, SCID , Phosphorylation , Prions/genetics , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...