73 results on '"Michael P. Blundell"'
Search Results
2. Tyrosine phosphorylation of WASP promotes calpain-mediated podosome disassembly
- Author
-
Lee Macpherson, James Monypenny, Michael P. Blundell, Giles O. Cory, Jessica Tomé-García, Adrian J. Thrasher, Gareth E. Jones, and Yolanda Calle
- Subjects
Diseases of the blood and blood-forming organs ,RC633-647.5 - Abstract
Podosomes are actin-based adhesions involved in migration of cells that have to cross tissue boundaries such as myeloid cells. The Wiskott Aldrich Syndrome Protein regulates de novo actin polymerization during podosome formation and it is cleaved by the protease calpain during podosome disassembly. The mechanisms that may induce the Wiskott Aldrich Syndrome Protein cleavage by calpain remain undetermined. We now report that in myeloid cells, tyrosine phosphorylation of the Wiskott Aldrich Syndrome Protein-tyrosine291 (Human)/tyrosine293 (mouse) not only enhances Wiskott Aldrich Syndrome Protein-mediated actin polymerization but also promotes its calpain-dependent degradation during podosome disassembly. We also show that activation of the Wiskott Aldrich Syndrome Protein leading to podosome formation occurs independently of tyrosine phosphorylation in spleen-derived dendritic cells. We conclude that tyrosine phosphorylation of the Wiskott Aldrich Syndrome Protein integrates dynamics of actin and cell adhesion proteins during podosome disassembly required for mobilization of myeloid cells during the immune response.
- Published
- 2012
- Full Text
- View/download PDF
3. Flow Cytometry as an Important Tool in Proteomic Profiling
- Author
-
Michael P, Blundell, Sharon L, Sanderson, and Tracey A, Long
- Subjects
Proteomics ,Humans ,Proteins ,Antigens ,Flow Cytometry ,Fluorescent Dyes ,Immunophenotyping - Abstract
Flow cytometry enables the simultaneous detection of multiple surface and intracellular antigens for proteomic profiling of cells. This allows characterization and identification of specific cell subtypes within a heterogeneous population and is usually called immunophenotyping. Antigen-specific antibodies, conjugated to various fluorophores, are incubated with the sample to identify each marker. Fluorescent light of various wavelengths can be separated, detected, and converted into a digital signal in a flow cytometer. Here we describe an eight-color experiment to identify key peripheral blood cell types; however, this technique can be expanded to detect more than 30 parameters simultaneously.
- Published
- 2021
4. Flow Cytometry as an Important Tool in Proteomic Profiling
- Author
-
Sharon L. Sanderson, Tracey A. Long, and Michael P. Blundell
- Subjects
0301 basic medicine ,Cell specific ,medicine.diagnostic_test ,Chemistry ,Proteomic Profiling ,Computational biology ,Flow cytometry ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immunophenotyping ,Fluorescent light ,Antigen ,030220 oncology & carcinogenesis ,medicine ,Peripheral blood cell ,Intracellular - Abstract
Flow cytometry enables the simultaneous detection of multiple surface and intracellular antigens for proteomic profiling of cells. This allows characterization and identification of specific cell subtypes within a heterogeneous population and is usually called immunophenotyping. Antigen-specific antibodies, conjugated to various fluorophores, are incubated with the sample to identify each marker. Fluorescent light of various wavelengths can be separated, detected, and converted into a digital signal in a flow cytometer. Here we describe an eight-color experiment to identify key peripheral blood cell types; however, this technique can be expanded to detect more than 30 parameters simultaneously.
- Published
- 2021
- Full Text
- View/download PDF
5. Natural killer cells differentiated in vitro from cord blood CD34 + cells are more advantageous for use as an immunotherapy than peripheral blood and cord blood natural killer cells
- Author
-
J. Alejandro Madrigal, Michael P. Blundell, Mark W. Lowdell, Aurore Saudemont, Anna Domogala, and Adrian J. Thrasher
- Subjects
Cancer Research ,Transplantation ,Lymphokine-activated killer cell ,Janus kinase 3 ,Immunology ,Cell Biology ,Biology ,Natural killer T cell ,03 medical and health sciences ,Interleukin 21 ,0302 clinical medicine ,Oncology ,NK-92 ,030220 oncology & carcinogenesis ,Cord blood ,Interleukin 12 ,Immunology and Allergy ,Cytotoxic T cell ,Genetics (clinical) ,030215 immunology - Abstract
Background aims Natural killer (NK) cells have the potential to become a successful immunotherapy as they can target malignant cells without being direct effectors of graft-versus-host disease. Our group has previously shown that large numbers of functional NK cells can be differentiated in vitro from umbilical cord blood (CB) CD34 + cells. To produce a clinically relevant and effective immunotherapy, we hypothesized that it is essential that the NK cells are able to proliferate and persist in vivo while maintaining an optimal activation status and killing capacity. Methods We evaluated the proliferation capacity, telomere length and terminal differentiation markers expressed by NK cells differentiated in vitro . We also determined how their cytotoxicity compared with peripheral blood (PB) NK cells and CBNK cells when targeting patient acute myeloid leukemia (AML) blasts and solid tumor cell lines. Results We found that the differentiated NK cells could respond to interleukin-2 and proliferate in vitro. Telomere length was significantly increased, whereas CD57 expression was significantly reduced compared with PBNK cells. The cytotoxicity of the differentiated NK cells was equivalent to that of the PBNK and CBNK cell controls, and priming consistently led to higher levels of killing of patient leukemic blasts and solid tumor cell lines in vitro . Interestingly, this activation step was not required to observe killing of patient AML blasts in vivo. Conclusion We are able to generate NK cells from CBCD34 + cells in high numbers, allowing for multiple infusions of highly cytotoxic NK cells that have potential to further proliferate in vivo , making them a desirable product for application as an immunotherapy in the clinic.
- Published
- 2017
- Full Text
- View/download PDF
6. Autoinflammatory periodic fever, immunodeficiency, and thrombocytopenia (PFIT) caused by mutation in actin-regulatory gene WDR1
- Author
-
Hannah E. Jones, Ariane Standing, Nigel Klein, Paul A. Brogan, Dale Moulding, Michael P. Blundell, Ebun Omoyinmi, Sira Nanthapisal, Sonia Melo Gomes, Horia Stanescu, Karolin Nowak, Ying Hong, Robert Kleta, Adrian J. Thrasher, Despina Eleftheriou, Kimberly Gilmour, Julien Record, Glenn Anderson, Alan Medlar, and Dessislava Malinova
- Subjects
0301 basic medicine ,Inflammasomes ,Immunology ,Caspase 1 ,Mutation, Missense ,NALP3 ,macromolecular substances ,Biology ,Pyrin domain ,03 medical and health sciences ,AIM2 ,Phagocytosis ,Mutant protein ,medicine ,Immunology and Allergy ,Missense mutation ,Humans ,Child ,Immunodeficiency ,Research Articles ,Hereditary Autoinflammatory Diseases ,Microfilament Proteins ,Brief Definitive Report ,Immunologic Deficiency Syndromes ,Interleukin-18 ,Inflammasome ,medicine.disease ,Thrombocytopenia ,Actins ,3. Good health ,030104 developmental biology ,biology.protein ,Female ,medicine.drug - Abstract
Standing et al. report a novel autoinflammatory disease caused by a homozygous missense mutation in the actin-regulating protein WDR1. The disease is characterized by periodic fevers, immunodeficiency, and thrombocytopenia, with increased polymerized actin in immune cells and increased IL-18 secretion., The importance of actin dynamics in the activation of the inflammasome is becoming increasingly apparent. IL-1β, which is activated by the inflammasome, is known to be central to the pathogenesis of many monogenic autoinflammatory diseases. However, evidence from an autoinflammatory murine model indicates that IL-18, the other cytokine triggered by inflammasome activity, is important in its own right. In this model, autoinflammation was caused by mutation in the actin regulatory gene WDR1. We report a homozygous missense mutation in WDR1 in two siblings causing periodic fevers with immunodeficiency and thrombocytopenia. We found impaired actin dynamics in patient immune cells. Patients had high serum levels of IL-18, without a corresponding increase in IL-18–binding protein or IL-1β, and their cells also secreted more IL-18 but not IL-1β in culture. We found increased caspase-1 cleavage within patient monocytes indicative of increased inflammasome activity. We transfected HEK293T cells with pyrin and wild-type and mutated WDR1. Mutant protein formed aggregates that appeared to accumulate pyrin; this could potentially precipitate inflammasome assembly. We have extended the findings from the mouse model to highlight the importance of WDR1 and actin regulation in the activation of the inflammasome, and in human autoinflammation.
- Published
- 2017
7. Molecular Evidence of Genome Editing in a Mouse Model of Immunodeficiency
- Author
-
Jianbin Wang, Chv Gan, Cynthia C. Bartholomae, F. J. Molina-Estevez, Christine Kinnon, Richard Gabriel, HH Abdul-Razak, Michael C. Holmes, V. Prakash, Celine J. Rocca, Rafael J. Yáñez-Muñoz, Philip D. Gregory, J Pantoglou, JA Bueren, M. E. Alonso-Ferrero, Adam Roberts, C von Kalle, Guillermo Guenechea, Steven J. Howe, Marina I. Garin, Adrian J. Thrasher, Michael P. Blundell, and Manfred Schmidt
- Subjects
0301 basic medicine ,lcsh:Medicine ,DNA-Activated Protein Kinase ,Mice, SCID ,Biology ,Genome ,Article ,Mice ,03 medical and health sciences ,Genome editing ,medicine ,Animals ,Humans ,lcsh:Science ,Immunodeficiency ,Gene Editing ,Severe combined immunodeficiency ,Multidisciplinary ,lcsh:R ,Nuclear Proteins ,medicine.disease ,Zinc finger nuclease ,3. Good health ,Transplantation ,Disease Models, Animal ,030104 developmental biology ,Cancer research ,Primary immunodeficiency ,Severe Combined Immunodeficiency ,lcsh:Q ,Ex vivo - Abstract
Genome editing is the introduction of directed modifications in the genome, a process boosted to therapeutic levels by designer nucleases. Building on the experience of ex vivo gene therapy for severe combined immunodeficiencies, it is likely that genome editing of haematopoietic stem/progenitor cells (HSPC) for correction of inherited blood diseases will be an early clinical application. We show molecular evidence of gene correction in a mouse model of primary immunodeficiency. In vitro experiments in DNA-dependent protein kinase catalytic subunit severe combined immunodeficiency (Prkdc scid) fibroblasts using designed zinc finger nucleases (ZFN) and a repair template demonstrated molecular and functional correction of the defect. Following transplantation of ex vivo gene-edited Prkdc scid HSPC, some of the recipient animals carried the expected genomic signature of ZFN-driven gene correction. In some primary and secondary transplant recipients we detected double-positive CD4/CD8 T-cells in thymus and single-positive T-cells in blood, but no other evidence of immune reconstitution. However, the leakiness of this model is a confounding factor for the interpretation of the possible T-cell reconstitution. Our results provide support for the feasibility of rescuing inherited blood disease by ex vivo genome editing followed by transplantation, and highlight some of the challenges.
- Published
- 2018
- Full Text
- View/download PDF
8. T-cell gene therapy for perforin deficiency corrects cytotoxicity defects and prevents hemophagocytic lymphohistiocytosis manifestations
- Author
-
Sujal, Ghosh, Marlene, Carmo, Miguel, Calero-Garcia, Ida, Ricciardelli, Juan Carlos, Bustamante Ogando, Michael P, Blundell, Axel, Schambach, Philip G, Ashton-Rickardt, Claire, Booth, Stephan, Ehl, Kai, Lehmberg, Adrian J, Thrasher, and H Bobby, Gaspar
- Subjects
Male ,WT, Wild-type ,T cells ,CD8-Positive T-Lymphocytes ,Lymphocytic Choriomeningitis ,Lymphohistiocytosis, Hemophagocytic ,Article ,TEM, Effector memory T ,HSCT, Hematopoietic stem cell transplantation ,Gene therapy ,FHL, Familial hemophagocytic lymphohistiocytosis ,NK, Natural killer ,TCM, Central memory T ,Cell Line, Tumor ,Animals ,Humans ,Lymphocytic choriomeningitis virus ,Mice, Knockout ,Perforin ,perforin deficiency ,Genetic Therapy ,IRES, Internal ribosomal entry site ,HLH, Hemophagocytic lymphohistiocytosis ,PRF1, Perforin 1 ,LCMV, Lymphocytic choriomeningitis virus ,Mice, Inbred C57BL ,hemophagocytic lymphohistiocytosis ,Child, Preschool ,GFP, Green fluorescent protein ,PS, Penicillin and streptomycin - Abstract
Background Mutations in the perforin 1 (PRF1) gene account for up to 58% of familial hemophagocytic lymphohistiocytosis syndromes. The resulting defects in effector cell cytotoxicity lead to hypercytokinemia and hyperactivation with inflammation in various organs. Objective We sought to determine whether autologous gene-corrected T cells can restore cytotoxic function, reduce disease activity, and prevent hemophagocytic lymphohistiocytosis (HLH) symptoms in in vivo models. Methods We developed a gammaretroviral vector to transduce murine CD8 T cells in the Prf−/− mouse model. To verify functional correction of Prf−/− CD8 T cells in vivo, we used a lymphocytic choriomeningitis virus (LCMV) epitope–transfected murine lung carcinoma cell tumor model. Furthermore, we challenged gene-corrected and uncorrected mice with LCMV. One patient sample was transduced with a PRF1-encoding lentiviral vector to study restoration of cytotoxicity in human cells. Results We demonstrated efficient engraftment and functional reconstitution of cytotoxicity after intravenous administration of gene-corrected Prf−/− CD8 T cells into Prf−/− mice. In the tumor model infusion of Prf−/− gene–corrected CD8 T cells eliminated the tumor as efficiently as transplantation of wild-type CD8 T cells. Similarly, mice reconstituted with gene-corrected Prf−/− CD8 T cells displayed complete protection from the HLH phenotype after infection with LCMV. Patients' cells showed correction of cytotoxicity in human CD8 T cells after transduction. Conclusion These data demonstrate the potential application of T-cell gene therapy in reconstituting cytotoxic function and protection against HLH in the setting of perforin deficiency.
- Published
- 2017
9. Towards gene therapy for EBV-associated posttransplant lymphoma with genetically modified EBV-specific cytotoxic T cells
- Author
-
Martin Pule, Adrian J. Thrasher, Persis Amrolia, Michael P. Blundell, Jennifer Brewin, and Ida Ricciardelli
- Subjects
Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,Adoptive cell transfer ,Lymphoma ,medicine.medical_treatment ,Genetic enhancement ,Calcineurin Inhibitors ,Immunology ,Drug Resistance ,chemical and pharmacologic phenomena ,Mice, SCID ,Biology ,medicine.disease_cause ,Immunotherapy, Adoptive ,Biochemistry ,Tacrolimus ,Mice ,Mice, Inbred NOD ,Transduction, Genetic ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Calcineurin ,hemic and immune systems ,Immunosuppression ,Genetic Therapy ,Gene Therapy ,Cell Biology ,Hematology ,medicine.disease ,Epstein–Barr virus ,surgical procedures, operative ,Mutation ,Genetic Engineering ,Immunosuppressive Agents ,T-Lymphocytes, Cytotoxic - Abstract
Epstein-Barr virus (EBV)-associated posttransplant lymphoma (PTLD) is a major cause of morbidity/mortality after hematopoietic stem cell (SCT) or solid organ (SOT) transplant. Adoptive immunotherapy with EBV-specific cytotoxic lymphocytes (CTLs), although effective in SCT, is less successful after SOT where lifelong immunosuppression therapy is necessary. We have genetically engineered EBV-CTLs to render them resistant to calcineurin (CN) inhibitor FK506 through retroviral transfer of a calcineurin A mutant (CNA12). Here we examined whether or not FK506-resistant EBV-CTLs control EBV-driven tumor progression in the presence of immunosuppression in a xenogeneic mouse model. NOD/SCID/IL2rγ(null) mice bearing human B-cell lymphoma were injected with autologous CTLs transduced with either CNA12 or eGFP in the presence/absence of FK506. Adoptive transfer of autologous CNA12-CTLs induced dramatic lymphoma regression despite the presence of FK506, whereas eGFP-CTLs did not. CNA12-CTLs persisted longer, homed to the tumor, and expanded more than eGFP-CTLs in mice treated with FK506. Mice receiving CNA12-CTLs and treated with FK506 survived significantly longer than control-treated animals. Our results demonstrate that CNA12-CTL induce regression of EBV-associated tumors in vivo despite ongoing immunosuppression. Clinical application of this novel approach may enhance the efficacy of adoptive transfer of EBV-CTL in SOT patients developing PTLD without the need for reduction in immunosuppressive therapy.
- Published
- 2014
- Full Text
- View/download PDF
10. Integrin linked kinase (ILK) regulates podosome maturation and stability in dendritic cells
- Author
-
Inés M. Antón, Gareth E. Jones, Manuel Rodríguez-Puyol, Inmaculada Bañón-Rodríguez, Michael P. Blundell, Ester Martín-Villar, Yolanda Calle, Mercedes Griera, Adrian J. Thrasher, Medical Research Council (UK), Ministerio de Ciencia e Innovación (España), Fundación Científica Asociación Española Contra el Cáncer, and Wellcome Trust
- Subjects
Male ,Podosome ,Integrin ,ComputingMilieux_LEGALASPECTSOFCOMPUTING ,WASP ,macromolecular substances ,Protein Serine-Threonine Kinases ,Transfection ,Cell Membrane Structures ,PI3K ,Biochemistry ,Filamentous actin ,Article ,Extracellular matrix ,Mice ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,0302 clinical medicine ,Cell Movement ,Extracellular ,Animals ,Humans ,Integrin-linked kinase ,ComputingMilieux_MISCELLANEOUS ,Actin ,030304 developmental biology ,0303 health sciences ,biology ,Wiskott–Aldrich syndrome protein ,Dendritic Cells ,Cell Biology ,Extracellular Matrix ,3. Good health ,Cell biology ,Mice, Inbred C57BL ,Data_GENERAL ,030220 oncology & carcinogenesis ,biology.protein ,ILK ,Wiskott-Aldrich Syndrome Protein ,Dendritic cell - Abstract
Under a Creative Commons license.-- et al., Podosomes are integrin-based adhesions fundamental for stabilisation of the leading lamellae in migrating dendritic cells (DCs) and for extracellular matrix (ECM) degradation. We have previously shown that soluble factors and chemokines such as SDF 1-α trigger podosome initiation whereas integrin ligands promote podosome maturation and stability in DCs. The exact intracellular signalling pathways that regulate the sequential organisation of podosomal components in response to extracellular cues remain largely undetermined. The Wiskott Aldrich Syndrome Protein (WASP) mediates actin polymerisation and the initial recruitment of integrins and associated proteins in a circular configuration surrounding the core of filamentous actin (F-actin) during podosome initiation. We have now identified integrin linked kinase (ILK) surrounding the podosomal actin core. We report that DC polarisation in response to chemokines and the assembly of actin cores during podosome initiation require PI3K-dependent clustering of the Wiskott Aldrich Syndrome Protein (WASP) in puncta independently of ILK. ILK is essential for the clustering of integrins and associated proteins leading to podosome maturation and stability that are required for degradation of the subjacent extracellular matrix and the invasive motility of DCs across connective tissue barriers. We conclude that WASP regulates DCs polarisation for migration and initiation of actin polymerisation downstream of PI3K in nascent podosomes. Subsequently, ILK mediates the accumulation of integrin-associated proteins during podosome maturation and stability for efficient degradation of the subjacent ECM during the invasive migration of DCs., This work has been supported by grants from the Burton Myeloma Programme (YC), Ministerio Español de Ciencia e Innovación (BFU2010-21374/BMC (IMA)), the Medical Research Council (G1100041, GEJ, IMA) Fundación Científica de la Asociación Española Contra el Cáncer (AECC) (EM-V and IB-R) and the Wellcome Trust (080373) (YC, MPB, AJT and GEJ).
- Published
- 2014
- Full Text
- View/download PDF
11. Publisher Correction: In Utero Gene Therapy (IUGT) Using GLOBE Lentiviral Vector Phenotypically Corrects the Heterozygous Humanised Mouse Model and Its Progress Can Be Monitored Using MRI Techniques
- Author
-
Simon N. Waddington, Panicos Shangaris, Durrgah L. Ramachandra, Daniel J. Stuckey, Wei Wang, Nahla Bakhamis, Adrian J. Thrasher, Simon Eaton, Ayad Eddaoudi, Anna L. David, S Subramaniam, Christina Flouri, Stavros P. Loukogeorgakis, Joy Archer, Paolo De Coppi, Michael Antoniou, Manfred G. Schmidt, Shanrun Liu, Laurence H. Jackson, Michael P. Blundell, Luca Urbani, Thomas M. Ryan, and Panagiotis Maghsoudlou
- Subjects
Mri techniques ,Pathology ,medicine.medical_specialty ,Multidisciplinary ,business.industry ,Genetic enhancement ,Science ,Viral vector ,In utero ,Medicine ,business - Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2019
12. Preclinical Demonstration of Lentiviral Vector-mediated Correction of Immunological and Metabolic Abnormalities in Models of Adenosine Deaminase Deficiency
- Author
-
Marlene Carmo, Xiaoyan Wang, Pei Yu Fu, H. Bobby Gaspar, Adrian J. Thrasher, Kenneth Cornetta, Neil J. Sebire, Michael L. Kaufman, Lin Zhang, Donald B. Kohn, Shantha Senadheera, Sabine Geiger, Roger P. Hollis, Denise A. Carbonaro, Aaron R. Cooper, Lynette D. Fairbanks, Rebecca Chan, Xiangyang Jin, Claudia Montiel-Equihua, Michael P. Blundell, and Arineh Sahaghian
- Subjects
Male ,Technology ,Adenosine Deaminase ,T-Lymphocytes ,Genetic enhancement ,Inbred C57BL ,Regenerative Medicine ,Medical and Health Sciences ,Mice ,Peptide Elongation Factor 1 ,Adenosine deaminase ,Stem Cell Research - Nonembryonic - Human ,Agammaglobulinemia ,Transduction, Genetic ,immune system diseases ,Drug Discovery ,2.1 Biological and endogenous factors ,Aetiology ,Promoter Regions, Genetic ,Cells, Cultured ,B-Lymphocytes ,Cultured ,hemic and immune systems ,Gene Therapy ,Biological Sciences ,3. Good health ,Molecular Medicine ,Stem Cell Research - Nonembryonic - Non-Human ,Female ,Original Article ,Development of treatments and therapeutic interventions ,Stem cell ,HT29 Cells ,Biotechnology ,Cells ,Virus Integration ,Genetic Vectors ,Biology ,Viral vector ,Promoter Regions ,Insertional mutagenesis ,Transduction ,Genetic ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Pharmacology ,Transplantation ,Severe combined immunodeficiency ,5.2 Cellular and gene therapies ,Animal ,Lentivirus ,Stem Cell Research ,medicine.disease ,Molecular biology ,Adenosine deaminase deficiency ,Mice, Inbred C57BL ,Disease Models, Animal ,HEK293 Cells ,Disease Models ,biology.protein ,Severe Combined Immunodeficiency - Abstract
Gene transfer into autologous hematopoietic stem cells by γ-retroviral vectors (gRV) is an effective treatment for adenosine deaminase (ADA)–deficient severe combined immunodeficiency (SCID). However, current gRV have significant potential for insertional mutagenesis as reported in clinical trials for other primary immunodeficiencies. To improve the efficacy and safety of ADA-SCID gene therapy (GT), we generated a self-inactivating lentiviral vector (LV) with a codon-optimized human cADA gene under the control of the short form elongation factor-1α promoter (LV EFS ADA). In ADA −/− mice, LV EFS ADA displayed high-efficiency gene transfer and sufficient ADA expression to rescue ADA −/− mice from their lethal phenotype with good thymic and peripheral T- and B-cell reconstitution. Human ADA-deficient CD34 + cells transduced with 1–5 × 10 7 TU/ml had 1–3 vector copies/cell and expressed 1–2x of normal endogenous levels of ADA, as assayed in vitro and by transplantation into immune-deficient mice. Importantly, in vitro immortalization assays demonstrated that LV EFS ADA had significantly less transformation potential compared to gRV vectors, and vector integration-site analysis by nrLAM-PCR of transduced human cells grown in immune-deficient mice showed no evidence of clonal skewing. These data demonstrated that the LV EFS ADA vector can effectively transfer the human ADA cDNA and promote immune and metabolic recovery, while reducing the potential for vector-mediated insertional mutagenesis.
- Published
- 2014
- Full Text
- View/download PDF
13. T-Cell Reconstitution after Thymus Xenotransplantation Induces Hair Depigmentation and Loss
- Author
-
Anna L. Furmanski, José Ignacio Saldaña, Michael P. Blundell, Adrian J. Thrasher, Ryan F.L. O'Shaughnessy, Neil J. Sebire, Tessa Crompton, and E. Graham Davies
- Subjects
CD4-Positive T-Lymphocytes ,Adoptive cell transfer ,Pathology ,medicine.medical_specialty ,T cell ,medicine.medical_treatment ,Transplantation, Heterologous ,Mice, Nude ,Thymus Gland ,Dermatology ,In Vitro Techniques ,Biology ,Biochemistry ,Mice ,030207 dermatology & venereal diseases ,03 medical and health sciences ,0302 clinical medicine ,Depigmentation ,Antigen ,medicine ,Animals ,Humans ,Hair Color ,Molecular Biology ,Cell Proliferation ,030304 developmental biology ,Homeodomain Proteins ,Mice, Knockout ,Autoimmune disease ,0303 health sciences ,Pigmentation ,Alopecia ,Cell Biology ,Hair follicle ,medicine.disease ,Adoptive Transfer ,Up-Regulation ,3. Good health ,Mice, Inbred C57BL ,Transplantation ,Disease Models, Animal ,medicine.anatomical_structure ,Thymus transplantation ,Melanocytes ,Original Article ,medicine.symptom ,Hair Follicle - Abstract
Here we present a mouse model for T-cell targeting of hair follicles, linking the pathogenesis of alopecia to that of depigmentation disorders. Clinically, thymus transplantation has been successfully used to treat T-cell immunodeficiency in congenital athymia, but is associated with autoimmunity. We established a mouse model of thymus transplantation by subcutaneously implanting human thymus tissue into athymic C57BL/6 nude mice. These xenografts supported mouse T-cell development. Surprisingly, we did not detect multiorgan autoimmune disease. However, in all transplanted mice, we noted a striking depigmentation and loss of hair follicles. Transfer of T cells from transplanted nudes to syngeneic black-coated RAG(-/-) recipients caused progressive, persistent coat-hair whitening, which preceded patchy hair loss in depigmented areas. Further transfer experiments revealed that these phenomena could be induced by CD4+ T cells alone. Immunofluorescent analysis suggested that Trp2+ melanocyte-lineage cells were decreased in depigmented hair follicles, and pathogenic T cells upregulated activation markers when exposed to C57BL/6 melanocytes in vitro, suggesting that these T cells are not tolerant to self-melanocyte antigens. Our data raise interesting questions about the mechanisms underlying tissue-specific tolerance to skin antigens.
- Published
- 2013
- Full Text
- View/download PDF
14. SAP gene transfer restores cellular and humoral immune function in a murine model of X-linked lymphoproliferative disease
- Author
-
Neil J. Sebire, Adrian J. Thrasher, Christine Rivat, Michael P. Blundell, Claire Booth, H. Bobby Gaspar, and M. E. Alonso-Ferrero
- Subjects
genetic structures ,Recombinant Fusion Proteins ,Genetic enhancement ,Green Fluorescent Proteins ,Immunology ,Plenary Paper ,Lymphoproliferative disorders ,Biology ,Biochemistry ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,medicine ,Animals ,Cell Lineage ,Signaling Lymphocytic Activation Molecule Associated Protein ,030304 developmental biology ,Mice, Knockout ,Immunity, Cellular ,0303 health sciences ,Reporter gene ,fungi ,Gene Transfer Techniques ,Hematopoietic Stem Cell Transplantation ,Intracellular Signaling Peptides and Proteins ,food and beverages ,Germinal center ,Hematopoietic stem cell ,X-linked lymphoproliferative disease ,Genetic Therapy ,Cell Biology ,Hematology ,biochemical phenomena, metabolism, and nutrition ,medicine.disease ,Molecular biology ,Lymphoproliferative Disorders ,eye diseases ,Immunity, Humoral ,3. Good health ,Killer Cells, Natural ,Transplantation ,Disease Models, Animal ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Natural Killer T-Cells - Abstract
X-linked lymphoproliferative disease (XLP1) arises from mutations in the gene encoding SLAM-associated protein (SAP) and leads to abnormalities of NKT-cell development, NK-cell cytotoxicity, and T-dependent humoral function. Curative treatment is limited to allogeneic hematopoietic stem cell (HSC) transplantation. We tested whether HSC gene therapy could correct the multilineage defects seen in SAP(-/-) mice. SAP(-/-) murine HSCs were transduced with lentiviral vectors containing either SAP or reporter gene before transplantation into irradiated recipients. NKT-cell development was significantly higher and NK-cell cytotoxicity restored to wild-type levels in mice receiving the SAP vector in comparison to control mice. Baseline immunoglobulin levels were significantly increased and T-dependent humoral responses to NP-CGG, including germinal center formation, were restored in SAP-transduced mice.We demonstrate for the first time that HSC gene transfer corrects the cellular and humoral defects in SAP(-/-) mice providing proof of concept for gene therapy in XLP1.
- Published
- 2013
- Full Text
- View/download PDF
15. Valproic Acid Confers Functional Pluripotency to Human Amniotic Fluid Stem Cells in a Transgene-free Approach
- Author
-
Joris Vermeesch, Sayandip Mukherjee, Gudrun E. Moore, Anthony Atala, Adrian J. Thrasher, Gemma N. Jones, Jennifer M. Frost, Nicholas M. Fisk, Michael P. Blundell, Pascale V. Guillot, Daniel Nettersheim, T Selvee Ramasamy, Kenneth Lay, Beata Nowakowska, Mara Cananzi, Dafni Moschidou, Paolo De Coppi, Hassan Abdulrazzak, Anju Phoolchund, James Adjaye, Mark H.F. Sullivan, Katharina Drews, and Hubert Schorle
- Subjects
Male ,Rex1 ,Induced Pluripotent Stem Cells ,Embryoid body ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Drug Discovery ,Genetics ,Humans ,Induced pluripotent stem cell ,Cell potency ,Molecular Biology ,030304 developmental biology ,Pharmacology ,0303 health sciences ,Induced stem cells ,Valproic Acid ,Amniotic Fluid ,Molecular biology ,Embryonic stem cell ,Cell biology ,Histone Deacetylase Inhibitors ,030220 oncology & carcinogenesis ,Molecular Medicine ,Original Article ,Female ,Stem cell ,Reprogramming - Abstract
Induced pluripotent stem cells (iPSCs) with potential for therapeutic applications can be derived from somatic cells via ectopic expression of a set of limited and defined transcription factors. However, due to risks of random integration of the reprogramming transgenes into the host genome, the low efficiency of the process, and the potential risk of virally induced tumorigenicity, alternative methods have been developed to generate pluripotent cells using nonintegrating systems, albeit with limited success. Here, we show that c-KIT+ human first-trimester amniotic fluid stem cells (AFSCs) can be fully reprogrammed to pluripotency without ectopic factors, by culture on Matrigel in human embryonic stem cell (hESC) medium supplemented with the histone deacetylase inhibitor (HDACi) valproic acid (VPA). The cells share 82% transcriptome identity with hESCs and are capable of forming embryoid bodies (EBs) in vitro and teratomas in vivo. After long-term expansion, they maintain genetic stability, protein level expression of key pluripotency factors, high cell-division kinetics, telomerase activity, repression of X-inactivation, and capacity to differentiate into lineages of the three germ layers, such as definitive endoderm, hepatocytes, bone, fat, cartilage, neurons, and oligodendrocytes. We conclude that AFSC can be utilized for cell banking of patient-specific pluripotent cells for potential applications in allogeneic cellular replacement therapies, pharmaceutical screening, and disease modeling. ispartof: Molecular Therapy vol:20 issue:10 pages:1953-1967 ispartof: location:United States status: published
- Published
- 2012
- Full Text
- View/download PDF
16. Engraftment defect of cytokine-cultured adult human mobilized CD34+ cells is related to reduced adhesion to bone marrow niche elements
- Author
-
Kwee Yong, Konstantina Kallinikou, Michael J. Watts, Fernando Anjos-Afonso, Michael P. Blundell, David C. Linch, Stuart J. Ings, Adrian J. Thrasher, and Dominique Bonnet
- Subjects
Adult ,Male ,CD34 ,Antigens, CD34 ,Mice, SCID ,Biology ,Rats, Sprague-Dawley ,Mice ,Colony-Stimulating Factors ,Bone Marrow ,Mice, Inbred NOD ,Cell Adhesion ,medicine ,Animals ,Humans ,Osteopontin ,Stem Cell Niche ,Progenitor cell ,Cell adhesion ,Cells, Cultured ,Graft Survival ,Hematopoietic Stem Cell Transplantation ,Hematology ,Hematopoietic Stem Cells ,Hematopoietic Stem Cell Mobilization ,Recombinant Proteins ,Rats ,Specific Pathogen-Free Organisms ,Cell biology ,Haematopoiesis ,medicine.anatomical_structure ,Immunology ,biology.protein ,Bone marrow ,Ex vivo ,Homing (hematopoietic) - Abstract
In vitro exposure of haematopoietic stem and progenitor cells (HSPC) to cytokines in expansion or gene therapy protocols reduces homing and engraftment in vivo. We have previously reported that this is related in part to altered tissue specificity of short-term homing, leading to loss of cells in non-haematopoietic tissues. Here we demonstrate that defective engraftment persists when cultured HSPC are transplanted by intrabone injection. Changes in engraftment function occur within 24 h of cytokine exposure, and are evident when engraftment is analysed solely in the injected bone. A novel ex vivo model of the bone marrow was developed, in which the attachment of infused HSPC in rodent long bones is reduced following culture with cytokines. Finally, cultured HSPC demonstrated reduced adhesion to N-cadherin, osteopontin and vascular cell-adhesion molecule-1, ligands present in bone marrow niches. These changes in adhesive function occur rapidly, and are not related to downregulation of the relevant receptors. Our findings suggest that cytokine exposure of adult human HSPC results in altered adhesion within bone marrow niches, further leading to reduced engraftment potential in vivo.
- Published
- 2012
- Full Text
- View/download PDF
17. The β-Globin Locus Control Region in Combination With the EF1α Short Promoter Allows Enhanced Lentiviral Vector-mediated Erythroid Gene Expression With Conserved Multilineage Activity
- Author
-
Aiste Monkeviciute, ME Alonso-Ferrero, Simone Scholz, Reiner Schulz, H. Bobby Gaspar, Marlene Carmo, Lin Zhang, Heba Saadeh, Manfred G. Schmidt, Yasuhiro Takeuchi, Michael Antoniou, Adrian J. Thrasher, Sean Knight, Lynette D. Fairbanks, Claudia Montiel-Equihua, Michael P. Blundell, and Mary Collins
- Subjects
Adenosine Deaminase ,Transgene ,Genetic enhancement ,Genetic Vectors ,Green Fluorescent Proteins ,beta-Globins ,Biology ,Viral vector ,Cell Line ,03 medical and health sciences ,Jurkat Cells ,Mice ,0302 clinical medicine ,Peptide Elongation Factor 1 ,Gene expression ,Drug Discovery ,Transcriptional regulation ,Genetics ,Animals ,Humans ,Promoter Regions, Genetic ,Molecular Biology ,Erythroid Precursor Cells ,Locus control region ,030304 developmental biology ,Regulation of gene expression ,Pharmacology ,0303 health sciences ,Lentivirus ,Genetic Therapy ,U937 Cells ,Hematopoietic Stem Cells ,Locus Control Region ,Peptide Elongation Factors ,Molecular biology ,Up-Regulation ,Lysosomal Storage Diseases ,Mice, Inbred C57BL ,HEK293 Cells ,Gene Expression Regulation ,Molecular Medicine ,Original Article ,030217 neurology & neurosurgery - Abstract
Some gene therapy strategies are compromised by the levels of gene expression required for therapeutic benefit, and also by the breadth of cell types that require correction. We designed a lentiviral vector system in which a transgene is under the transcriptional control of the short form of constitutively acting elongation factor 1α promoter (EFS) combined with essential elements of the locus control region of the β-globin gene (β-LCR). We show that the β-LCR can upregulate EFS activity specifically in erythroid cells but does not alter EFS activity in myeloid or lymphoid cells. Experiments using the green fluorescent protein (GFP) reporter or the human adenosine deaminase (ADA) gene demonstrate 3–7 times upregulation in vitro but >20 times erythroid-specific upregulation in vivo, the effects of which were sustained for 1 year. The addition of the β-LCR did not alter the mutagenic potential of the vector in in vitro mutagenesis (IM) assays although microarray analysis showed that the β-LCR upregulates ~9% of neighboring genes. This vector design therefore combines the benefits of multilineage gene expression with high-level erythroid expression, and has considerable potential for correction of multisystem diseases including certain lysosomal storage diseases through a hematopoietic stem cell (HSC) gene therapy approach.
- Published
- 2012
- Full Text
- View/download PDF
18. British Society for Gene Therapy 2012March 9, 2012UCL Institute of Child Health, London
- Author
-
Simone Scholz, Michael Antoniou, H. Bobby Gaspar, Aiste Monkeviciute, Marlene Carmo, ME Alonso-Ferrero, Heba Saadeh, Mary Collins, Yasuhiro Takeuchi, Lin Zhang, Adrian J. Thrasher, Reiner Schulz, Sean Knight, Manfred G. Schmidt, Claudia Montiel-Equihua, Lynette D. Fairbanks, and Michael P. Blundell
- Subjects
Expression (architecture) ,Genetics ,Molecular Medicine ,Physiology ,Biology ,Molecular Biology ,Gene ,Cell biology - Published
- 2012
- Full Text
- View/download PDF
19. Biochemical Correction of X-CGD by a Novel Chimeric Promoter Regulating High Levels of Transgene Expression in Myeloid Cells
- Author
-
Manuel Grez, Harry L. Malech, Adrian J. Thrasher, Christine Kinnon, Giorgia Santilli, C Beilin, Elena Almarza, Michael P. Blundell, Kathryn L. Parsley, Juan A. Bueren, Christian Brendel, Uimook Choi, and Sneha Haria
- Subjects
Cathepsin G ,Myeloid ,Genetic enhancement ,CAAT box ,Granulomatous Disease, Chronic ,Mice ,0302 clinical medicine ,Genes, X-Linked ,hemic and lymphatic diseases ,Drug Discovery ,Myeloid Cells ,Transgenes ,Receptors, Immunologic ,Promoter Regions, Genetic ,Spleen Focus-Forming Viruses ,Regulation of gene expression ,0303 health sciences ,Stem Cells ,Cell Differentiation ,3. Good health ,Haematopoiesis ,Enhancer Elements, Genetic ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Molecular Medicine ,Original Article ,DNA Copy Number Variations ,Recombinant Fusion Proteins ,Transgene ,Genetic Vectors ,Molecular Sequence Data ,Biology ,Viral vector ,03 medical and health sciences ,Cell Line, Tumor ,Proto-Oncogene Proteins ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,030304 developmental biology ,Pharmacology ,Reporter gene ,Binding Sites ,Base Sequence ,Lentivirus ,Terminal Repeat Sequences ,NADPH Oxidases ,Genetic Therapy ,Hematopoietic Stem Cells ,Molecular biology ,Retroviridae ,Gene Expression Regulation ,Proto-Oncogene Proteins c-fes ,Mutagenesis ,CCAAT-Enhancer-Binding Proteins ,Trans-Activators ,Granulocytes - Abstract
X-linked chronic granulomatous disease (X-CGD) is a primary immunodeficiency caused by mutations in the CYBB gene encoding the phagocyte nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase catalytic subunit gp91(phox). A recent clinical trial for X-CGD using a spleen focus-forming virus (SFFV)-based γ-retroviral vector has demonstrated clear therapeutic benefits in several patients although complicated by enhancer-mediated mutagenesis and diminution of effectiveness over time due to silencing of the viral long terminal repeat (LTR). To improve safety and efficacy, we have designed a lentiviral vector that directs transgene expression primarily in myeloid cells. To this end, we created a synthetic chimeric promoter that contains binding sites for myeloid transcription factors CAAT box enhancer-binding family proteins (C/EBPs) and PU.1, which are highly expressed during granulocytic differentiation. As predicted, the chimeric promoter regulated higher reporter gene expression in myeloid than in nonmyeloid cells, and in human hematopoietic progenitors upon granulocytic differentiation. In a murine model of stem cell gene therapy for X-CGD, the chimeric vector resulted in high levels of gp91(phox) expression in committed myeloid cells and granulocytes, and restored normal NADPH-oxidase activity. These findings were recapitulated in human neutrophils derived from transduced X-CGD CD34(+) cells in vivo, and suggest that the chimeric promoter will have utility for gene therapy of myeloid lineage disorders such as CGD.
- Published
- 2011
- Full Text
- View/download PDF
20. Correction of Murine SCID-X1 by Lentiviral Gene Therapy Using a Codon-optimized IL2RG Gene and Minimal Pretransplant Conditioning
- Author
-
Axel Schambach, Niek P. van Til, Shazia Arshad, Michael P. Blundell, Ali Nowrouzi, Adrian J. Thrasher, Claudia Cattoglio, Fang Zhang, Trudi P. Visser, Christof von Kalle, Gerard Wagemaker, Marshall W. Huston, Monique M.A. Verstegen, Yuedan Li, Fulvio Mavilio, Martijn H. Brugman, Manfred Schmidt, Christopher Baum, and Hematology
- Subjects
Genetic enhancement ,T-Lymphocytes ,Receptors, Antigen, T-Cell ,Mice, SCID ,Biology ,SCID ,Viral vector ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Antigen ,Receptors ,Drug Discovery ,Genetics ,medicine ,Animals ,Codon ,Molecular Biology ,Gene ,030304 developmental biology ,Pharmacology ,0303 health sciences ,Severe combined immunodeficiency ,Phosphoglycerate kinase ,B-Lymphocytes ,Drug Discovery3003 Pharmaceutical Science ,Lentivirus ,Hematopoietic Stem Cell Transplantation ,Original Articles ,Genetic Therapy ,T-Cell ,medicine.disease ,Virology ,Molecular biology ,3. Good health ,030220 oncology & carcinogenesis ,Antibody Formation ,Molecular Medicine ,Severe Combined Immunodeficiency ,Interleukin Receptor Common gamma Subunit ,Ex vivo - Abstract
Clinical trials have demonstrated the potential of ex vivo hematopoietic stem cell gene therapy to treat X-linked severe combined immunodeficiency (SCID-X1) using retroviral vectors, leading to immune system functionality in the majority of treated patients without pretransplant conditioning. The success was tempered by insertional oncogenesis in a proportion of the patients. To reduce the genotoxicity risk, a self-inactivating (SIN) lentiviral vector (LV) with improved expression of a codon optimized human interleukin-2 receptor gamma gene (IL2RG) cDNA (co gamma c), regulated by its 1.1 kb promoter region (gamma cPr), was compared in efficacy to the viral spleen focus forming virus (SF) and the cellular phosphoglycerate kinase (PGK) promoters. Pretransplant conditioning of IL2rg(-/-) mice resulted in long-term reconstitution of T and B lymphocytes, normalized natural antibody titers, humoral immune responses, ConA/IL-2 stimulated spleen cell proliferation, and polyclonal T-cell receptor gene rearrangements with a clear integration preference of the SF vector for proto-oncogenes, contrary to the PGK and gamma cPr vectors. We conclude that SIN lentiviral gene therapy using co gamma c driven by the gamma cPr or PGK promoter corrects the SCID phenotype, potentially with an improved safety profile, and that low-dose conditioning proved essential for immune competence, allowing for a reduced threshold of cell numbers required. Received 23 February 2011; accepted 31 May 2011; published online 12 July 2011. doi:10.1038/mt.2011.127
- Published
- 2011
- Full Text
- View/download PDF
21. The Wiskott-Aldrich Syndrome: The Actin Cytoskeleton and Immune Cell Function
- Author
-
Michael P. Blundell, Austen Worth, Gerben Bouma, and Adrian J. Thrasher
- Subjects
0303 health sciences ,lcsh:R5-920 ,Biochemistry (medical) ,Clinical Biochemistry ,General Medicine ,macromolecular substances ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,hemic and lymphatic diseases ,Genetics ,lcsh:Medicine (General) ,Molecular Biology ,030304 developmental biology - Abstract
Wiskott-Aldrich syndrome (WAS) is a rare X-linked recessive primary immunodeficiency characterised by immune dysregulation, microthrombocytopaenia, eczema and lymphoid malignancies. Mutations in the WAS gene can lead to distinct syndrome variations which largely, although not exclusively, depend upon the mutation. Premature termination and deletions abrogate Wiskott-Aldrich syndrome protein (WASp) expression and lead to severe disease (WAS). Missense mutations usually result in reduced protein expression and the phenotypically milder X-linked thrombocytopenia (XLT) or attenuated WAS [1-3]. More recently however novel activating mutations have been described that give rise to X-linked neutropenia (XLN), a third syndrome defined by neutropenia with variable myelodysplasia [4-6]. WASP is key in transducing signals from the cell surface to the actin cytoskeleton, and a lack of WASp results in cytoskeletal defects that compromise multiple aspects of normal cellular activity including proliferation, phagocytosis, immune synapse formation, adhesion and directed migration.
- Published
- 2010
22. Phosphorylation of WASp is a key regulator of activity and stability in vivo
- Author
-
Gerben Bouma, Siobhan O. Burns, Giles O. Cory, Michael P. Blundell, Gareth E. Jones, Samuel Mirando, Christine Kinnon, Lisa S. Westerberg, Austen Worth, Yolanda Calle, Adrian J. Thrasher, Scott B. Snapper, Joao Metelo, Dale Moulding, and Lucy A. Cowell
- Subjects
Mutant ,Regulator ,Mice, Transgenic ,macromolecular substances ,In Vitro Techniques ,Biology ,medicine.disease_cause ,Cell Line ,Mice ,chemistry.chemical_compound ,Phagocytosis ,Cell Movement ,Chlorocebus aethiops ,medicine ,Animals ,Humans ,Phosphorylation ,Tyrosine ,Mice, Knockout ,Mutation ,Binding Sites ,Multidisciplinary ,Tyrosine phosphorylation ,Chemotaxis ,Biological Sciences ,Actins ,Recombinant Proteins ,Hematopoiesis ,Wiskott-Aldrich Syndrome ,Mice, Inbred C57BL ,Amino Acid Substitution ,Proteasome ,chemistry ,Biochemistry ,COS Cells ,Mutagenesis, Site-Directed ,Wiskott-Aldrich Syndrome Protein - Abstract
The Wiskott-Aldrich syndrome protein (WASp) is a key cytoskeletal regulator in hematopoietic cells. Covalent modification of a conserved tyrosine by phosphorylation has emerged as an important potential determinant of activity, although the physiological significance remains uncertain. In a murine knockin model, mutation resulting in inability to phosphorylate Y293 (Y293F) mimicked many features of complete WASp-deficiency. Although a phosphomimicking mutant Y293E conferred enhanced actin-polymerization, the cellular phenotype was similar due to functional dysregulation. Furthermore, steady-state levels of Y293E-WASp were markedly reduced compared to wild-type WASp and Y293F-WASp, although partially recoverable by treatment of cells with proteasome inhibitors. Consequently, tyrosine phosphorylation plays a critical role in normal activation of WASp in vivo, and is indispensible for multiple tasks including proliferation, phagocytosis, chemotaxis, and assembly of adhesion structures. Furthermore, it may target WASp for proteasome-mediated degradation, thereby providing a default mechanism for self-limiting stimulation of the Arp2/3 complex.
- Published
- 2009
- Full Text
- View/download PDF
23. Hematopoietic-Specific Lentiviral Vectors Circumvent Cellular Toxicity Due to Ectopic Expression of Wiskott-Aldrich Syndrome Protein
- Author
-
Miguel G. Toscano, Enrique García-Olivares, Michael P. Blundell, Francisco Martin, Adrian J. Thrasher, Cecilia Frecha, Ignacio J. Molina, Marién Cobo, and Karim Benabdellah
- Subjects
Transcription, Genetic ,Wiskott–Aldrich syndrome ,Hematopoietic System ,Recombinant Fusion Proteins ,T-Lymphocytes ,Genetic enhancement ,Genetic Vectors ,Green Fluorescent Proteins ,macromolecular substances ,Biology ,Cell Line ,Mice ,Genetics ,medicine ,Animals ,Humans ,Progenitor cell ,Molecular Biology ,Cytoskeleton ,Cell Proliferation ,Severe combined immunodeficiency ,Cell Death ,Lentivirus ,Wiskott–Aldrich syndrome protein ,Genetic Therapy ,medicine.disease ,Virology ,Wiskott-Aldrich Syndrome ,Cell biology ,Haematopoiesis ,Organ Specificity ,biology.protein ,Molecular Medicine ,Ectopic expression ,Stem cell ,Wiskott-Aldrich Syndrome Protein - Abstract
Efficient and safe gene modification of hematopoietic stem cells is a requirement for gene therapy of primary immunodeficiencies such as Wiskott-Aldrich syndrome. However, deregulated expression or ectopic expression in the progeny of transduced nonhematopoietic progenitor cells may lead to unwanted toxicity. We therefore analyzed the effect of ectopic expression of Wiskott-Aldrich syndrome protein (WASp) and the potential benefits of hematopoietic-specific lentiviral vectors (driven by the WAS proximal promoter). Overexpression of WASp by constitutive lentiviral vectors is highly toxic in nonhematopoietic cells because it causes dramatic changes in actin localization and polymerization that result in decreased cell viability, as evidenced by a significant growth disadvantage of WASp-overexpressing nonhematopoietic cells and increased cell death. These toxic effects do not affect cells of hematopoietic origin because, remarkably, we found that WASp cannot be readily overexpressed in T cells, even after multiple vector integrations per cell. The adverse cellular effects found after transduction of nonhematopoietic cells with constitutive lentiviral vectors are overcome by the use of transcriptionally targeted lentiviral vectors expressing WASp, which, at the same time, are efficient tools for gene therapy of WAS as demonstrated by their ability to reconstitute cellular defects from WASp-deficient mouse and human cells. We therefore postulate that transcriptionally regulated lentiviral vectors represent a safer and efficient alternative for the development of clinical protocols of WAS gene therapy.
- Published
- 2008
- Full Text
- View/download PDF
24. Regulatory T cells inhibit CD34+ cell differentiation into NK cells by blocking their proliferation
- Author
-
Michael P. Blundell, Martha Luevano, Aurore Saudemont, Divya K. Shah, Adrian J. Thrasher, Alejandro Madrigal, Anna Domogala, Isabela Pedroza-Pacheco, and Nicola Jackson
- Subjects
0301 basic medicine ,Adoptive cell transfer ,Cell ,chemical and pharmacologic phenomena ,Biology ,T-Lymphocytes, Regulatory ,Article ,Cell therapy ,03 medical and health sciences ,Interleukin 21 ,Mice ,medicine ,Animals ,Humans ,Cell Proliferation ,Multidisciplinary ,Cell growth ,Hematopoietic stem cell ,hemic and immune systems ,medicine.disease ,Adoptive Transfer ,Hematopoiesis ,Killer Cells, Natural ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,Graft-versus-host disease ,Immunology ,Cancer research - Abstract
Graft versus Host Disease (GvHD) remains one of the main complications after hematopoietic stem cell transplantation (HSCT). Due to their ability to suppress effector cells, regulatory T cells (Tregs) have been proposed as a cellular therapy to prevent GvHD, however they also inhibit the functions of natural killer (NK) cells, key effectors of the Graft versus Leukemia effect. In this study, we have explored whether a Tregs therapy will also impact on NK cell differentiation. Using an in vitro model of hematopoietic stem cell (HSC) differentiation into NK cells, we found that activated Tregs led to a 90% reduction in NK cell numbers when added at the time of commitment to the NK cell lineage. This effect was contact dependent and was reversible upon Tregs depletion. The few NK cells that developed in these cultures were mature and exhibited normal functions. Furthermore, adoptive transfer of activated Tregs in rag-/- γc-/- mice abrogated HSC differentiation into NK cells thus confirming our in vitro findings. Collectively, these results demonstrate for the first time that activated Tregs can inhibit NK cell differentiation from HSC under specific conditions.
- Published
- 2016
- Full Text
- View/download PDF
25. Stable Gene Transfer to Muscle Using Non-integrating Lentiviral Vectors
- Author
-
Nicola J. Philpott, Michael P. Blundell, L Apolonia, Simon N. Waddington, Gerben Bouma, Carolina Fernandes, Mary Collins, Adrian J. Thrasher, and NJ Ward
- Subjects
Transgene ,Genetic Vectors ,Gene Expression ,Genome, Viral ,Gene delivery ,Genome ,Cell Line ,Insertional mutagenesis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Transduction, Genetic ,Gene expression ,Drug Discovery ,Genetics ,Gene silencing ,Animals ,Humans ,Transgenes ,Molecular Biology ,030304 developmental biology ,Pharmacology ,0303 health sciences ,biology ,Integrases ,Muscles ,Lentivirus ,Long terminal repeat ,3. Good health ,Integrase ,Cell biology ,030220 oncology & carcinogenesis ,DNA, Viral ,Mutation ,biology.protein ,Molecular Medicine - Abstract
Human immunodeficiency virus (HIV)-based lentiviral vectors (LVs) hold immense promise for gene delivery applications because of their relatively large packaging capacity and their ability to infect a range of cell types. The genome of HIV non-specifically integrates into the host genome, and this promotes efficient, stable transgene expression in dividing cells. However, integration can also be problematic because of variations in gene expression among cells, possible gene silencing and, most importantly, insertional mutagenesis which can lead to undesirable effects such as malignant transformation. In order to alleviate these problems, we have developed a range of non-integrating LVs (NILVs) by introducing point mutations into the catalytic site, chromosome binding site, and viral DNA binding site of the viral integrase (IN). In addition, we have mutated the IN attachment (att) sites within the HIV long terminal repeats (LTRs). All of the vectors produced show efficient reverse transcription and transgene expression in dividing cells and prolonged expression in non-dividing myotubes. Finally, we show that NILV can be used for achieving highly effective gene transfer and expression in muscle in vivo.
- Published
- 2007
- Full Text
- View/download PDF
26. Severe autoinflammatory disease caused by mutation in a gene controlling actin cytoskeletal dynamics and cure with allogeneic haematopoetic stem cell transplantation
- Author
-
Ariane Standing, Nigel Klein, Michael P. Blundell, Ebun Omoyinmi, S Melo Gomes, H Jones, Julien Record, Dale Moulding, Despina Eleftheriou, Dessislava Malinova, Paul A. Brogan, K Nowak, Adrian J. Thrasher, Sira Nanthapisal, and Ying Hong
- Subjects
Mutation ,animal diseases ,chemical and pharmacologic phenomena ,macromolecular substances ,biochemical phenomena, metabolism, and nutrition ,Immune dysregulation ,Biology ,medicine.disease_cause ,Actin cytoskeleton ,medicine.disease ,Cell biology ,Transplantation ,Immune system ,Rheumatology ,Pediatrics, Perinatology and Child Health ,Immunology ,medicine ,Oral Presentation ,bacteria ,Immunology and Allergy ,Pediatrics, Perinatology, and Child Health ,Stem cell ,Cytoskeleton ,Immunodeficiency - Abstract
The actin cytoskeleton is crucial at many junctures of normal immune function, and consequently there are many immune specific regulators of actin dynamics. A growing number of primary immunodeficiencies are being defined as caused by mutations in the genes encoding these regulators. In addition to immunodeficiency, immune dysregulation and autoinflammation are increasingly recognised to arise from defects within this pathway.
- Published
- 2015
- Full Text
- View/download PDF
27. Deficiency in the Wiskott-Aldrich protein induces premature proplatelet formation and platelet production in the bone marrow compartment
- Author
-
Abdelali Jalil, Adlen Foudi, Sabine Charrier, Brigitte Franc, Siham Boukour, William Vainchenker, Elisabeth M. Cramer, Najet Debili, Fawzia Louache, Adrian J. Thrasher, Richard W. Farndale, Siham Sabri, Martine Jandrot-Perrus, and Michael P. Blundell
- Subjects
Blood Platelets ,Podosome ,Immunology ,Wiskott-Aldrich Syndrome Protein, Neuronal ,macromolecular substances ,Biology ,Biochemistry ,Collagen Type I ,Collagen receptor ,Mice ,Bone Marrow ,medicine ,Animals ,Platelet ,Thrombopoiesis ,Receptor ,Wiskott–Aldrich syndrome protein ,Cell Differentiation ,Cell Biology ,Hematology ,Thrombocytopenia ,Mice, Inbred C57BL ,Disease Models, Animal ,medicine.anatomical_structure ,Cancer research ,biology.protein ,Bone marrow ,Integrin alpha2beta1 ,GPVI ,Megakaryocytes - Abstract
The pathophysiology of microthrombocytopenia in the Wiskott-Aldrich syndrome (WAS) and its milder form, X-linked thrombocytopenia (XLT), is unclear. Although quantitative defects are correctable by splenectomy, residual platelet abnormalities are suggestive of intrinsic disturbances of production. In contrast to human patients, murine models of WASp deficiency exhibit only mild thrombocytopenia, and platelets are of normal size. Here, we have identified a critical role for WASp during murine platelet biogenesis. By electron microscopy, WASp-deficient MKs appeared to have shed platelets ectopically within the bone marrow space. WASp-deficient megakaryocytes (MKs) also displayed defects in response to fibrillar collagen I (CI) in vitro, the major matrix component of bone. These included a loss of normal CI receptor (α2β1 integrin)-mediated inhibition of proplatelet formation, a marked abrogation of SDF-1-induced chemotactic migration of CD41+ MKs adherent to CI, and an almost complete lack of actin-rich podosomes, normally induced by interaction between CI and its receptors GPVI or α2β1 integrin. These findings highlight the central and highly specialized role of WASp in MKs during platelet biogenesis, and may provide a mechanism for the mild thrombocytopenia observed in WASp-deficient mice. In addition, they suggest a novel explanation for some of the platelet abnormalities characteristic of patients with WAS. (Blood. 2006;108:134-140)
- Published
- 2006
- Full Text
- View/download PDF
28. Coordinated oncogenic transformation and inhibition of host immune responses by the PAX3-FKHR fusion oncoprotein
- Author
-
Stephen Nabarro, Antigoni Papanastasiou, Neil J. Sebire, Nourredine Himoudi, Adrian J. Thrasher, Sian Gibson, Mike Hubank, Glenda Canderan, Kimberly Gilmour, John Anderson, and Michael P. Blundell
- Subjects
STAT3 Transcription Factor ,Oncogene Proteins, Fusion ,Transcription, Genetic ,Immunology ,Down-Regulation ,Biology ,Major histocompatibility complex ,Article ,Cell Line ,Mice ,Immune system ,Cell Line, Tumor ,Immune Tolerance ,Animals ,Humans ,Paired Box Transcription Factors ,Immunology and Allergy ,Transcription factor ,Mice, Knockout ,Fusion protein ,Up-Regulation ,Cell biology ,Mice, Inbred C57BL ,Cell Transformation, Neoplastic ,STAT protein ,biology.protein ,Inflammation Mediators ,Signal transduction ,Janus kinase - Abstract
Tumors have evolved elaborate mechanisms for evading immune detection, such as production of immunoinhibitory cytokines and down-regulation of major histocompatibility complex (MHC) expression. We have studied PAX3-FKHR as an example of an oncogenic fusion protein associated with an aggressive metastatic cancer. We show that PAX3-FKHR alters expression of genes that are normally regulated by Janus kinase/signal transducer and activator of transcription (STAT) signaling pathways. This occurs as a result of a specific interaction between PAX3-FKHR and the STAT3 transcription factor, which results in a dramatic reduction in tumor MHC expression, and an alteration in local cytokine concentrations to inhibit surrounding inflammatory cells and immune detection. Collectively, these data show that an oncogenic transcription factor can promote tumor growth and tissue invasion while inhibiting local inflammatory and immune responses. This is the first time that an immunomodulatory role has been described for an oncogenic fusion protein.
- Published
- 2005
- Full Text
- View/download PDF
29. Impaired dendritic-cell homing in vivo in the absence of Wiskott-Aldrich syndrome protein
- Author
-
Christine Kinnon, Samantha J. Hardy, Adrian J. Thrasher, Michael P. Blundell, Joanna Sinclair, Gareth E. Jones, Jörg Zwirner, Oliver Schulz, David R. Katz, Jessica Strid, and Sofia de Noronha
- Subjects
Time Factors ,Langerhans cell ,T-Lymphocytes ,Immunology ,Bone Marrow Cells ,macromolecular substances ,Dermatitis, Contact ,Biochemistry ,Mice ,chemistry.chemical_compound ,Cell Movement ,medicine ,Animals ,Fluorescein isothiocyanate ,Antigen-presenting cell ,Skin ,Mice, Knockout ,Chemokine CCL21 ,biology ,Wiskott–Aldrich syndrome protein ,Oxazolone ,Proteins ,Cell migration ,Dendritic Cells ,Cell Biology ,Hematology ,Dendritic cell ,Actin cytoskeleton ,Wiskott-Aldrich Syndrome ,Cell biology ,Disease Models, Animal ,medicine.anatomical_structure ,chemistry ,Chemokines, CC ,Langerhans Cells ,biology.protein ,Lymph Nodes ,Spleen ,Wiskott-Aldrich Syndrome Protein ,Homing (hematopoietic) - Abstract
Regulated migration and spatial localization of dendritic cells (DCs) are critical events during the initiation of physiologic immune responses and maintenance of tolerance. Here we have used cells deficient in the Wiskott-Aldrich syndrome protein (WASp) to demonstrate the importance of dynamic remodeling of the actin cytoskeleton for these trafficking processes to occur in vitro and in vivo. On fibronectin-coated surfaces, WASp-null immature murine DCs exhibited defects both of attachment and detachment, resulting in impaired net translocation compared with normal cells. The chemokinetic response to CCL21, which is critical for normal lymphatic trafficking, was also abrogated in the absence of WASp. In vivo in both fluorescein isothiocyanate (FITC) and oxazolone contact hypersensitivity models, WASp-null Langerhans cell (LC) migration was compromised, as judged by exit from the skin as well as by homing to the draining lymph node (LN). Furthermore, following systemic challenge with lipopolysaccharide (LPS) or toxoplasma-derived antigen, WASp-null DCs showed incomplete redistribution to T-cell areas in the spleen. Instead, they were retained ectopically in the marginal zone. DC trafficking in vivo is therefore dependent on a normally regulated actin cytoskeleton, which performs an essential function during maintenance of physiologic immunity and when disturbed may contribute significantly to the immunopathology of Wiskott-Aldrich Syndrome.
- Published
- 2005
- Full Text
- View/download PDF
30. Perforin gene transfer into hematopoietic stem cells improves immune dysregulation in murine models of perforin deficiency
- Author
-
Marlene, Carmo, Kimberly A, Risma, Paritha, Arumugam, Swati, Tiwari, Adrianne E, Hontz, Claudia A, Montiel-Equihua, Maria E, Alonso-Ferrero, Michael P, Blundell, Axel, Schambach, Christopher, Baum, Punam, Malik, Adrian J, Thrasher, Michael B, Jordan, and H Bobby, Gaspar
- Subjects
Killer Cells, Natural ,Disease Models, Animal ,Mice ,Phenotype ,Perforin ,Gene Transfer Techniques ,Animals ,Mice, Transgenic ,Original Article ,CD8-Positive T-Lymphocytes ,Hematopoietic Stem Cells ,Lymphohistiocytosis, Hemophagocytic - Abstract
Defects in perforin lead to the failure of T and NK cell cytotoxicity, hypercytokinemia, and the immune dysregulatory condition known as familial hemophagocytic lymphohistiocytosis (FHL). The only curative treatment is allogeneic hematopoietic stem cell transplantation which carries substantial risks. We used lentiviral vectors (LV) expressing the human perforin gene, under the transcriptional control of the ubiquitous phosphoglycerate kinase promoter or a lineage-specific perforin promoter, to correct the defect in different murine models. Following LV-mediated gene transfer into progenitor cells from perforin-deficient mice, we observed perforin expression in mature T and NK cells, and there was no evidence of progenitor cell toxicity when transplanted into irradiated recipients. The resulting perforin-reconstituted NK cells showed partial recovery of cytotoxicity, and we observed full recovery of cytotoxicity in polyclonal CD8(+) T cells. Furthermore, reconstituted T cells with defined antigen specificity displayed normal cytotoxic function against peptide-loaded targets. Reconstituted CD8(+) lymphoblasts had reduced interferon-γ secretion following stimulation in vitro, suggesting restoration of normal immune regulation. Finally, upon viral challenge, mice with30% engraftment of gene-modified cells exhibited reduction of cytokine hypersecretion and cytopenias. This study demonstrates the potential of hematopoietic stem cell gene therapy as a curative treatment for perforin-deficient FHL.
- Published
- 2014
31. Trisomy 21 mid-trimester amniotic fluid induced pluripotent stem cells maintain genetic signatures during reprogramming: implications for disease modeling and cryobanking
- Author
-
Assunta Pandolfi, Panicos Shangaris, Drew Ellershaw, Anna L. David, Jonathan J. Waters, Pascale V. Guillot, Paolo De Coppi, Sayandip Mukherjee, Adrian J. Thrasher, Agostino Pierro, Steven W. Shaw, Caterina Pipino, Michael P. Blundell, Marina Cavazzana, Peggy Sung, and Gustavo Mostoslavsky
- Subjects
Cryopreservation ,Induced Pluripotent Stem Cells ,Amniotic stem cells ,Cell Biology ,Embryoid body ,Biology ,Amniotic Fluid ,Embryonic stem cell ,Models, Biological ,Mice ,SOX2 ,Pregnancy ,Amniotic epithelial cells ,Prenatal Diagnosis ,Immunology ,Cancer research ,Animals ,Humans ,Female ,Stem cell ,Down Syndrome ,Induced pluripotent stem cell ,Reprogramming ,Developmental Biology ,Biotechnology - Abstract
Trisomy 21 is the most common chromosomal abnormality and is associated primarily with cardiovascular, hematological, and neurological complications. A robust patient-derived cellular model is necessary to investigate the pathophysiology of the syndrome because current animal models are limited and access to tissues from affected individuals is ethically challenging. We aimed to derive induced pluripotent stem cells (iPSCs) from trisomy 21 human mid-trimester amniotic fluid stem cells (AFSCs) and describe their hematopoietic and neurological characteristics. Human AFSCs collected from women undergoing prenatal diagnosis were selected for c-KIT(+) and transduced with a Cre-lox-inducible polycistronic lentiviral vector encoding SOX2, OCT4, KLF-4, and c-MYC (50,000 cells at a multiplicity of infection (MOI) 1-5 for 72 h). The embryonic stem cell (ESC)-like properties of the AFSC-derived iPSCs were established in vitro by embryoid body formation and in vivo by teratoma formation in RAG2(-/-), γ-chain(-/-), C2(-/-) immunodeficient mice. Reprogrammed cells retained their cytogenetic signatures and differentiated into specialized hematopoietic and neural precursors detected by morphological assessment, immunostaining, and RT-PCR. Additionally, the iPSCs expressed all pluripotency markers upon multiple rounds of freeze-thawing. These findings are important in establishing a patient-specific cellular platform of trisomy 21 to study the pathophysiology of the aneuploidy and for future drug discovery.
- Published
- 2014
32. Sheep CD34+ amniotic fluid cells have hematopoietic potential and engraft after autologous in utero transplantation
- Author
-
Anna L. David, Po-Jen Cheng, Graça Almeida-Porada, Christopher D. Porada, Paolo De Coppi, Durrgah L. Ramachandra, Michael P. Blundell, S. W. Steven Shaw, Adrian J. Thrasher, Fanos Georgiades, Caterina Pipino, M Boyd, Panicos Shangaris, and Panagiotis Maghsoudlou
- Subjects
Pathology ,medicine.medical_specialty ,Transplantation, Heterologous ,CD34 ,Cell- and Tissue-Based Therapy ,Antigens, CD34 ,Mice, SCID ,Biology ,Transplantation, Autologous ,In utero transplantation ,Cell therapy ,Mice ,Fetus ,Mice, Inbred NOD ,Pregnancy ,medicine ,Autologous transplantation ,Animals ,Sheep ,Hematopoietic Stem Cell Transplantation ,Cell Biology ,Amniotic Fluid ,Transplantation ,Haematopoiesis ,medicine.anatomical_structure ,Immunology ,Molecular Medicine ,Female ,Bone marrow ,Stem cell ,Developmental Biology - Abstract
Unmatched allogeneic in utero stem cell transplantation (IUSCT) produces poor engraftment unless the fetus has congenital immunodeficiency, probably because of maternal and fetal immune responses to injected cells. We studied the functional hematopoietic potential of transduced green fluorescent protein (GFP+) sheep amniotic fluid (AF) stem cells, before and after autologous IUSCT. CD34+ cells were selected from first trimester sheep AF, transduced overnight, and injected intravenously into NOD-SCID-gamma (NSG) mice. At 3 months, primary recipient bone marrow (BM) was injected into secondary NSG recipients. GFP+ cells were detected in the hematopoietic organs and peripheral blood of primary and secondary recipients at 3 months. Autologous IUSCT (transduced GFP+CD34+AF) was performed in fetal sheep. Six months postnatally, lamb BM was injected into secondary NSG recipients. GFP+ cells were detected in the peripheral blood of primary and secondary recipients. This confirms the hematopoietic potential of AF stem cells supporting the concept of autologous IUSCT to treat congenital hematopoietic disease. Stem Cells 2015;33:122–132
- Published
- 2014
33. Cutting Edge: The Wiskott-Aldrich Syndrome Protein Is Required for Efficient Phagocytosis of Apoptotic Cells
- Author
-
Michael P. Blundell, Yann Leverrier, Paul M. Brickell, Christine Kinnon, Adrian J. Thrasher, Anne J. Ridley, and Roberto Lorenzi
- Subjects
Phagocytic cup ,Phagocytosis ,Immunology ,Apoptosis ,Bone Marrow Cells ,Mice, Inbred Strains ,RAC1 ,Jurkat cells ,Cell Line ,Proinflammatory cytokine ,Apoptotic cell clearance ,Jurkat Cells ,Mice ,Animals ,Humans ,Immunology and Allergy ,Cells, Cultured ,Mice, Knockout ,biology ,Wiskott–Aldrich syndrome protein ,Proteins ,Opsonin Proteins ,Wiskott-Aldrich Syndrome ,Cell biology ,Cell culture ,Immunoglobulin G ,Macrophages, Peritoneal ,biology.protein ,Female ,Wiskott-Aldrich Syndrome Protein - Abstract
Phagocytosis of apoptotic cells by macrophages and dendritic cells is necessary for clearance of proinflammatory debris and for presentation of viral, tumor, and self Ags. While a number of receptors involved in the cognate recognition of apoptotic cells by phagocytes have been identified, the signaling events that result in internalization remain poorly understood. Here we demonstrate that clearance of apoptotic cells is accompanied by recruitment of the Wiskott-Aldrich syndrome (WAS) protein to the phagocytic cup and that it’s absence results in delayed phagocytosis both in vitro and in vivo. Therefore, we propose that WAS protein plays an important and nonredundant role in the safe removal of apoptotic cells and that deficiency contributes significantly to the immune dysregulation of WAS. The efficiency of apoptotic cell clearance may be a key determinant in the suppression of tissue inflammation and prevention of autoimmunity.
- Published
- 2001
- Full Text
- View/download PDF
34. A Defined Window for Efficient Gene Marking of Severe Combined Immunodeficient-Repopulating Cells Using a Gibbon Ape Leukemia Virus-Pseudotyped Retroviral Vector
- Author
-
Jacki P. Goldman, Manuel Grez, Gaby Brouns, Roland J. Levinsky, Christophe Demaison, Christine Kinnon, Adrian J. Thrasher, and Michael P. Blundell
- Subjects
Genetic Markers ,Myeloid ,Genetic enhancement ,Genetic Vectors ,Population ,Antigens, CD34 ,Mice, SCID ,Biology ,Viral vector ,Mice ,Mice, Inbred NOD ,Genetics ,medicine ,Animals ,Humans ,Progenitor cell ,education ,Molecular Biology ,Interleukin 3 ,Reporter gene ,education.field_of_study ,Flow Cytometry ,Virology ,Molecular biology ,Haematopoiesis ,medicine.anatomical_structure ,Leukemia Virus, Gibbon Ape ,Cytokines ,Molecular Medicine ,Severe Combined Immunodeficiency - Abstract
We have investigated the minimal time required for efficient transduction of human hematopoietic repopulating cells using a surrogate nonobese diabetic (NOD)/severe combined immunodeficient (SCID) xenoengraftment assay. Cord blood CD34+ cells were transduced to high levels over 24-48 hr in the presence of Flt-3 ligand, stem cell factor, interleukin 3, and interleukin 6. Under these conditions, high levels of NOD/SCID repopulating activity were preserved, but the levels of gene marking in engrafting cell populations measured by expression of a reporter transgene were low. Extension of the transduction period by 24 hr (total culture period, 72 hr) under the same cytokine conditions resulted in high levels of gene marking, but on closer analysis expression was limited predominantly to the myeloid population. Efficient transduction of both lymphoid and myeloid lineages could be achieved only if the transduction protocol was extended by a further 24 hr (total culture period, 96 hr), suggesting that myeloid lineage-committed precursors are capable of repopulation, and that over shorter time periods transduction is largely restricted to this population. This adds to the emerging evidence of heterogeneity within the SRC compartment, and has important implications for the interpretation of this assay in stem cell transplantation and gene transfer studies.
- Published
- 2000
- Full Text
- View/download PDF
35. Enhanced human cell engraftment in mice deficient in RAG2 and the common cytokine receptor γ chain
- Author
-
Michael P. Blundell, J P Di Santo, Christine Kinnon, L Lopes, Adrian J. Thrasher, and Jacki P. Goldman
- Subjects
Severe combined immunodeficiency ,medicine.medical_treatment ,Lymphocyte ,Hematology ,T lymphocyte ,Biology ,medicine.disease ,Natural killer cell ,Immune tolerance ,medicine.anatomical_structure ,Cytokine ,Immune system ,Immunology ,medicine ,CD8 - Abstract
Xenotransplantation of human cells into immunodeficient mice has been used to develop models of human haemopoiesis and lymphoid cell function. However, the utility of existing mouse strains can be limited by shortened life-spans, spontaneous production of functional lymphocytes with ageing, and residual innate immunity leading to variable levels of engraftment. Mice with a deletion of the common cytokine receptor gamma chain (gamma c) gene have reduced numbers of peripheral T and B lymphocytes, and absent natural killer cell (NK) activity. A genetic cross with a recombinase activating gene 2 (RAG2)-deficient strain produced mice doubly homozygous for the gamma c and RAG2 null alleles (gamma c-/RAG2-). These mice have a stable phenotype characterized by the absence of all T lymphocyte. B lymphocyte and NK cell function. Injection of human B-lymphoblastoid cells resulted in earlier fatal metastatic lymphoproliferative disease than in NOD/LtSz-scid controls. This was particularly evident in animals injected intravenously, possibly because of residual NK activity in NOD/LtSz-scid mice. Levels of engraftment with peripheral-blood-derived human lymphocytes were also increased and associated with higher CD4/CD8 ratios. These findings demonstrate that this new strain of immunodeficient mice has significant advantages over existing strains for engraftment of human cells, and may be useful for study of adoptive immunotherapy and novel therapies for GvHD and HIV infection.
- Published
- 1998
- Full Text
- View/download PDF
36. Wiskott-Aldrich syndrome protein-deficient hematopoietic cells can be efficiently mobilized by granulocyte colony-stimulating factor
- Author
-
Sabine Charrier, Anne Galy, Adrian J. Thrasher, Gregory Cédrone, Michael P. Blundell, William Vainchenker, Fawzia Louache, Généthon, Immunologie moléculaire et biothérapies innovantes (IMBI), École Pratique des Hautes Études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université d'Évry-Val-d'Essonne (UEVE)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Généthon, Laboratoire Environnements Sédimentaires - Géosciences Marines (GM/LES), Institut Français de Recherche pour l'Exploitation de la Mer (IFREMER), Institut Gustave Roussy (IGR), Hématopoïèse normale et pathologique (U1170 Inserm), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Paris-Sud - Paris 11 (UP11)-Institut Gustave Roussy (IGR), Hématopoïèse normale et pathologique, Université Paris-Sud - Paris 11 (UP11)-Institut Gustave Roussy (IGR)-Institut National de la Santé et de la Recherche Médicale (INSERM), École pratique des hautes études (EPHE), Laboratoire Environnements Sédimentaires (LES), Géosciences Marines (GM), and Institut Français de Recherche pour l'Exploitation de la Mer (IFREMER)-Institut Français de Recherche pour l'Exploitation de la Mer (IFREMER)
- Subjects
Male ,Mice, 129 Strain ,[SDV]Life Sciences [q-bio] ,Biology ,CXCR4 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Granulocyte Colony-Stimulating Factor ,medicine ,Animals ,Humans ,Progenitor cell ,Hematopoietic Stem Cell Mobilization ,030304 developmental biology ,Interleukin 3 ,Mice, Knockout ,0303 health sciences ,Hematology ,Hematopoietic Stem Cells ,Recombinant Proteins ,Wiskott-Aldrich Syndrome ,3. Good health ,Endothelial stem cell ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Immunology ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Bone marrow ,Original Articles and Brief Reports ,Stem cell ,Wiskott-Aldrich Syndrome Protein ,Homing (hematopoietic) - Abstract
International audience; The Wiskott-Aldrich syndrome protein is an essential cytoskeleton regulator found in cells of the hematopoietic lineage and controls the motility of leukocytes. The impact of WAS gene deficiency on the mobilization of hematopoietic progenitor/stem cells in circulation has remained unexplored but information would be pertinent in the context of autologous gene therapy of Wiskott-Aldrich syndrome. The response to granulocyte-colony stimulating factor mobilization was investigated in a murine WAS knock-out model of the disease, by measuring hematologic parameters, circulation and engraftment of hematopoietic progenitor/stem cells. In the steady-state, adult WAS knock-out mice have B-cell lymphopenia, marked neutrophilia, increased counts of circulating hematopoietic progenitor cells and splenomegaly, presumably caused by the retention of hematopoietic progenitor cells due to high levels of splenic CXCL12. In spite of these anomalies, the administration of granulocyte-colony-stimulating factor mobilizes progenitor/stem cells in WAS knock-out mice to the same level and with the same kinetics as in wild-type control mice. Mobilized peripheral blood cells from WAS knock-out mice can be transduced and are able to engraft into lethally-irradiated hosts reconstituting multiple lineages of cells and providing more effective radio-protection than mobilized cells from wild-type control mice. Surprisingly, the homing and the peripheral blood recovery of B lymphocytes was influenced by the background of the host. Thus, in the absence of Wiskott-Aldrich syndrome protein, effective mobilization is achieved but partial correction may occur as a result of an abnormal hematopoietic environment.
- Published
- 2013
- Full Text
- View/download PDF
37. Megakaryocytes assemble podosomes that degrade matrix and protrude through basement membrane
- Author
-
James Monypenny, Christian Gachet, Milica Vukovic, Amy Sinclair, Catherine Léon, Hannah Schachtner, Adrian J. Thrasher, Laura M. Machesky, Gareth E. Jones, Steve P. Watson, Michael P. Blundell, Alison M. Michie, Simon D. J. Calaminus, Steven G. Thomas, and Tessa L. Holyoake
- Subjects
Blood Platelets ,Podosome ,Immunology ,Biology ,Biochemistry ,Basement Membrane ,Extracellular matrix ,Mice ,Megakaryocyte ,medicine ,Extracellular ,Animals ,Humans ,Cytoskeleton ,Actin ,Cells, Cultured ,Basement membrane ,Mice, Knockout ,Myosin Type II ,Infant, Newborn ,Fibrinogen ,Cell Biology ,Hematology ,Matrix Metalloproteinases ,Cell biology ,Extracellular Matrix ,Mice, Inbred C57BL ,medicine.anatomical_structure ,HEK293 Cells ,Invadopodia ,Cell Surface Extensions ,Megakaryocytes - Abstract
Megakaryocytes give rise to platelets via extension of proplatelet arms, which are released through the vascular sinusoids into the bloodstream. Megakaryocytes and their precursors undergo varying interactions with the extracellular environment in the bone marrow during their maturation and positioning in the vascular niche. We demonstrate that podosomes are abundant in primary murine megakaryocytes adherent on multiple extracellular matrix substrates, including native basement membrane. Megakaryocyte podosome lifetime and density, but not podosome size, are dependent on the type of matrix, with podosome lifetime dramatically increased on collagen fibers compared with fibrinogen. Podosome stability and dynamics depend on actin cytoskeletal dynamics but not matrix metalloproteases. However, podosomes degrade matrix and appear to be important for megakaryocytes to extend protrusions across a native basement membrane. We thus demonstrate for the first time a fundamental requirement for podosomes in megakaryocyte process extension across a basement membrane, and our results suggest that podosomes may have a role in proplatelet arm extension or penetration of basement membrane.
- Published
- 2013
38. Feeder-Free Derivation of Functional Platelets from Human Induced Pluripotent Stem Cells
- Author
-
ip Mukherjee, Pei-Ju Sung, Adrian J. Thrasher, Say, and Michael P. Blundell
- Subjects
Agonist ,medicine.drug_class ,Cell culture ,Platelet disorder ,Immunology ,medicine ,Fibrinogen binding ,Platelet ,Thrombopoiesis ,Biology ,Induced pluripotent stem cell ,In vitro ,Cell biology - Abstract
Derivation of patient specific induced pluripotent stem cells, in combination with directed platelet differentiation, provides a promising platform to study human thrombopoiesis or platelet disorders, and has clear potential for future clinical applications. However, efficient differentiation of human induced pluripotent stem cells (iPSCs) into functional blood cells remains challenging. In this study, we report a novel protocol for the generation of CD41a+, CD42b+, and CD61+ functional platelets from human iPSCs under feeder-free culture conditions. Platelets derived from this feeder-free system showed similar fibrinogen binding activity after agonist stimulation when compared to platelets derived from a previously reported cell line co culture system. Evidence showed these culture-derived platelets responded to different agonist stimulation by increased expression of activation markers (CD62P and PAC1) as expected. Together, these results provide an important step towards generating in vitro functional platelets from an unlimited source of patient-specific induced pluripotent stem cells using an animal component-free culture system.
- Published
- 2013
- Full Text
- View/download PDF
39. Human Amniotic Fluid Stem Cells Have Hematopoietic Potential In Vivo
- Author
-
Durrgah L. Ramachandra, Paolo De Coppi, Sindhu Subramaniam, Marlene Carmo, Michael P. Blundell, Alan W. Flake, Eleni Antoniadou, Ania Manson, Stavros P. Loukogeorgakis, Steven J. Howe, Alfonso M Tedeschi, Anna L. David, and Panicos Shangaris
- Subjects
business.industry ,medicine.medical_treatment ,Immunology ,Hematopoietic Tissue ,CD34 ,Cell Biology ,Hematology ,Hematopoietic stem cell transplantation ,Biochemistry ,Andrology ,Transplantation ,Haematopoiesis ,medicine.anatomical_structure ,Cord blood ,medicine ,Bone marrow ,Stem cell ,business - Abstract
Introduction Human hematopoietic stem cell transplantation (HSCT) has been used to treat a range of hematological and immunological disorders. As a result, the demand for hematopoietic stem cells (HSC) in clinical applications is increasing. Amniotic fluid stem cells (AFSC) serve as a potential alternative cell source for therapy. Amniotic fluid can be derived by amniocentesis or therapeutic amniodrainage. AFSC are multi-potent, have low risk of tumorigenicity, can be expanded and do not have legal or ethical limitations. The significant hematopoietic activity of murine AFSC led us to explore the potential of human AFSC (CD117/c-Kit+) towards hematopoietic differentiation and to reconstitution in vivo. Methods Human AFSC (2nd and 3rd trimester) and cord blood HSC (CB-HSC; control) were selected for CD117 and CD34 respectively using a MoFlo XDP sorter. Sorted cells (104 in 200μl PBS) were injected intravenously into sub-lethally irradiated NOD-SCID/IL2rγnull (NSG) mice (n=6/group). Hematopoietic engraftment of human cells (% of human CD45+ within total CD45+) and multi-lineage reconstitution (myeloid: CD13, CD14, CD15 and lymphoid: CD3, CD4 and CD8) were assessed at 16 weeks in blood, bone marrow (BM) and spleen by flow cytometry. For subsequent secondary transplants, BM mononuclear cells (MNC) derived from BM harvested from primary recipients of mice were intravenously injected into secondary recipients (1.5x107 MNC in 200μl PBS). Hematopoietic engraftment was assessed at 16 weeks post-transplantation (n=6/group). For further analysis of human donor cell engraftment, Q-PCR was performed on spleen samples harvested from primary and secondary recipients using oligonucleotide primers specific for human ALU repeat sequences; Immunohistochemistry was carried out using anti-human CD45 antibody and detected with a commercially available kit (Dako EnVision Plus, Dako). Results are expressed as mean±SEM, and statistical analysis was performed using 1-way ANOVA with Bonferroni post-hoc tests. Results Human AFSC engrafted the hematopoietic system of NSG mice at levels similar to the ones achieved with CB-HSC (blood: AFSC 7.5±1.3% vs. CB-HSC 6.1±2.2%, p=0.6; BM: AFSC 46.3±7.9% vs. CB-HSC 38.3±8.2%, p=0.6; spleen: AFSC 39.6±9.3% vs. CB-HSC 34.7±10.5%, p=0.7). Similarly, at 16 weeks following secondary transplantation, human donor cell engraftment was comparable between groups in blood (AFSC 11.5 ± 3.9% vs. CB-HSC 16.9 ± 3.9%, p=0.3) and other hematopoietic tissues. Q-PCR and immunohistochemistry confirmed donor cell engraftment in AFSC and CB-HSC groups. Importantly, there were no differences between groups in multi-lineage differentiation at 16 weeks post primary and secondary transplantation. Conclusion Human CD117/c-Kit+ AFSC have functional, multi-lineage hematopoietic potential that is similar to the current "gold-standard" stem cell source for hematopoietic transplantation. The ease of isolation during early gestation, as well as their gene-engineering and expansion potential make human AFSC a novel autologous fetal cell source for pre- and post-natal therapy of inherited hematological disorders. Disclosures No relevant conflicts of interest to declare.
- Published
- 2016
- Full Text
- View/download PDF
40. Animal Models of Human Primary Immunodeficiency Diseases
- Author
-
Michael P. Blundell and Christine Kinnon
- Subjects
Genetics ,Immune system ,Primary immunodeficiency ,medicine ,Gene targeting ,Disease ,Computational biology ,Biology ,Acquired immune system ,medicine.disease ,Embryonic stem cell ,Genome ,Phenotype - Abstract
Animals with immunodeficiencies provide useful model systems for the study of corresponding immunological disorders in man. There has been substantial progress made in the availability and numbers of animal models since the 1970s when naturally occurring models were the only source. Improved molecular biology techniques, embryonic stem (ES) cell technologies and the sequencing of whole animal genomes has not only increased the number of models but also has led to improved models, particularly in mice. The increased understanding of the immune system, immune disorders and the generation of novel therapies owes much to animal models of disease. Further work is now in generating larger animal models which in addition to expressing the immunological disorders have a more directly comparable physiology. Key Concepts: Animal models of human immunodeficiency have been useful in studying the immune system. The phenotypes resulting from genetic defects in animals do not always mirror those seen in humans. Technologies exist to knock-out, knock-in and knock-down relevant genes as well as replace them with human loci in animal models to improve the comparability of the phenotype. Murine models are the most commonly described and used animal models but technologies are being developed for larger animal models. Animal models have been crucial in the development of novel therapies for immunodeficiencies. Keywords: immunodeficient; animal models; X-linked; inherited disease; innate and adaptive immune system; knock-out and knock-in mice; gene targeting; spontaneous mutation
- Published
- 2012
- Full Text
- View/download PDF
41. Human mid-trimester amniotic fluid stem cells cultured under embryonic stem cell conditions with Valproic acid acquire pluripotent characteristics
- Author
-
Anthony Atala, Dafni Moschidou, Michael P. Blundell, Pascale V. Guillot, Adrian J. Thrasher, Sayandip Mukherjee, Gemma N. Jones, Nicholas M. Fisk, and Paolo De Coppi
- Subjects
Pluripotent Stem Cells ,Homeobox protein NANOG ,Kruppel-Like Transcription Factors ,Gene Expression ,Embryoid body ,Biology ,Proto-Oncogene Proteins c-myc ,Kruppel-Like Factor 4 ,Mice ,SOX2 ,Pregnancy ,Animals ,Humans ,Cell Shape ,Cells, Cultured ,Embryonic Stem Cells ,reproductive and urinary physiology ,Cell Proliferation ,SOXB1 Transcription Factors ,Valproic Acid ,Teratoma ,Mesenchymal Stem Cells ,Amniotic stem cells ,Neoplasms, Experimental ,Cell Biology ,Hematology ,Telomere ,Amniotic Fluid ,Antigens, Differentiation ,Embryonic stem cell ,Culture Media ,Up-Regulation ,Cell biology ,Histone Deacetylase Inhibitors ,Pregnancy Trimester, Second ,Amniotic epithelial cells ,embryonic structures ,Immunology ,Female ,biological phenomena, cell phenomena, and immunity ,Stem cell ,Octamer Transcription Factor-3 ,Reprogramming ,Developmental Biology - Abstract
Human mid-trimester amniotic fluid stem cells (AFSC) have promising applications in regenerative medicine, being broadly multipotent with an intermediate phenotype between embryonic (ES) and mesenchymal stem cells (MSC). Despite this propluripotent phenotype, AFSC are usually cultured in adherence in a serum-based expansion medium, and how expansion in conditions sustaining pluripotency might affect their phenotype remains unknown. We recently showed that early AFSC from first trimester amniotic fluid, which endogenously express Sox2 and Klf4, can be reprogrammed to pluripotency without viral vectors using the histone deacetylase inhibitor valproic acid (VPA). Here, we show that mid-trimester AFSC cultured under MSC conditions contained a subset of cells endogenously expressing telomerase, CD24, OCT4, C-MYC, and SSEA4, but low/null levels of SOX2, NANOG, KLF4, SSEA3, TRA-1-60, and TRA-1-81, with cells unable to form embryoid bodies (EBs) or teratomas. In contrast, AFSC cultured under human ESC conditions were smaller in size, grew faster, formed colonies, upregulated OCT4 and C-MYC, and expressed KLF4 and SOX2, but not NANOG, SSEA3, TRA-1-60, and TRA-1-81. Supplementation with VPA for 5 days further upregulated OCT4, KLF4, and SOX2, and induced expression of NANOG, SSEA3, TRA-1-60, and TRA-1-81, with cells now able to form EBs and teratomas. We conclude that human mid-trimester AFSC, which may be isolated autologously during pregnancy without ethics restriction, can acquire pluripotent characteristics without the use of ectopic factors. Our data suggest that this medium-dependant approach to pluripotent mid-trimester AFSC reflects true reprogramming and not the selection of prepluripotent cells.
- Published
- 2012
42. Functional human artificial chromosomes are generated and stably maintained in human embryonic stem cells
- Author
-
Zoia L. Monaco, Adrian J. Thrasher, Mohammed Yusuf, Sayandip Mukherjee, Emanuela V. Volpi, Daniela Moralli, Mohammad A. Mandegar, Suhail Khoja, William James, David Yiu Leung Chan, Michael P. Blundell, and Sally A. Cowley
- Subjects
Neurons ,Cell division ,Cell growth ,Fluorescent Antibody Technique ,Cell Differentiation ,Articles ,Herpesvirus 1, Human ,General Medicine ,Human artificial chromosome ,Biology ,Amplicon ,Flow Cytometry ,Embryonic stem cell ,Molecular biology ,Chromosomes, Artificial, Human ,Cell Line ,Cell culture ,Genetics ,Humans ,Stem cell ,Molecular Biology ,Gene ,Embryonic Stem Cells ,Genetics (clinical) - Abstract
We present a novel and efficient non-integrating gene expression system in human embryonic stem cells (hESc) utilizing human artificial chromosomes (HAC), which behave as autonomous endogenous host chromosomes and segregate correctly during cell division. HAC are important vectors for investigating the organization and structure of the kinetochore, and gene complementation. HAC have so far been obtained in immortalized or tumour-derived cell lines, but never in stem cells, thus limiting their potential therapeutic application. In this work, we modified the herpes simplex virus type 1 amplicon system for efficient transfer of HAC DNA into two hESc. The deriving stable clones generated green fluorescent protein gene-expressing HAC at high frequency, which were stably maintained without selection for 3 months. Importantly, no integration of the HAC DNA was observed in the hESc lines, compared with the fibrosarcoma-derived control cells, where the exogenous DNA frequently integrated in the host genome. The hESc retained pluripotency, differentiation and teratoma formation capabilities. This is the first report of successfully generating gene expressing de novo HAC in hESc, and is a significant step towards the genetic manipulation of stem cells and potential therapeutic applications.
- Published
- 2011
43. Cytoskeletal remodeling mediated by WASp in dendritic cells is necessary for normal immune synapse formation and T-cell priming
- Author
-
Michael P. Blundell, Siobhan O. Burns, Ariadna Mendoza-Naranjo, Elena De Falco, Kathryn L. Parsley, Adrian J. Thrasher, and Gerben Bouma
- Subjects
Immunological Synapses ,Ovalbumin ,Wiskott–Aldrich syndrome ,Recombinant Fusion Proteins ,T-Lymphocytes ,T cell ,Immunology ,Receptors, Antigen, T-Cell ,Priming (immunology) ,Mice, Transgenic ,Biology ,Lymphocyte Activation ,Biochemistry ,Immunological synapse ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Bacterial Proteins ,Cell Movement ,Genes, Reporter ,medicine ,Animals ,Humans ,Calcium Signaling ,Cytoskeleton ,Antigen-presenting cell ,Crosses, Genetic ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Genetic Complementation Test ,Microtubule organizing center ,Dendritic Cells ,Cell Biology ,Hematology ,medicine.disease ,Wiskott-Aldrich Syndrome ,Cell biology ,Mice, Inbred C57BL ,Luminescent Proteins ,medicine.anatomical_structure ,Interleukin 12 ,Wiskott-Aldrich Syndrome Protein ,030215 immunology - Abstract
Rearrangement of the cytoskeleton in T cells plays a critical role in the organization of a complex signaling interface referred to as immunologic synapse (IS). Surprisingly, the contribution of antigen presenting cells, in particular dendritic cells (DCs), to the structure and function of the IS has not been investigated in as much detail. We have used a natural model of cytoskeletal dysfunction caused by deficiency of the Wiskott-Aldrich syndrome protein (WASp) to explore the contribution of the DC cytoskeleton to IS formation and to T-cell priming. In an antigen-specific system, T-DC contacts were found to be less stable when DCs alone lacked WASp, and associated with multiple defects of IS structure. As a consequence, DCs were unable to support normal IL-12 secretion, and events downstream of TCR signaling were abrogated, including increased calcium flux, microtubule organizing center (MTOC) polarization, phosphorylation of ZAP-70, and T-cell proliferation. Formation of an effective signaling interface is therefore dependent on active cytoskeletal rearrangements in DCs even when T cells are functionally competent. Deficiency of DC-mediated activities may contribute significantly to the varied immunodysregulation observed in patients with WAS, and also in those with limited myeloid reconstitution after allogeneic hematopoietic stem cell transplantation.
- Published
- 2011
44. Lentivirus-mediated Reprogramming of Somatic Cells in the Absence of Transgenic Transcription Factors
- Author
-
Sayandip Mukherjee, Nicole M. Kane, Joanne C. Mountford, Michael P. Blundell, Jenny A. Greig, Wai K. Lee, Adrian J. Thrasher, Manfred Schmidt, Andrew H. Baker, Christof von Kalle, Miles D. Houslay, Graeme Milligan, and Ali Nowrouzi
- Subjects
Somatic cell ,Transgene ,Induced Pluripotent Stem Cells ,Biology ,03 medical and health sciences ,0302 clinical medicine ,microRNA ,Gene expression ,Drug Discovery ,Genetics ,Humans ,Induced pluripotent stem cell ,Transcription factor ,Molecular Biology ,030304 developmental biology ,Pharmacology ,0303 health sciences ,Cell Differentiation ,Cell biology ,Gene expression profiling ,Retroviridae ,030220 oncology & carcinogenesis ,Molecular Medicine ,Original Article ,Reprogramming - Abstract
Retroviral vectors remain the most efficient and widely applied system for induction of pluripotency. However, mutagenic effects have been documented in both laboratory and clinical gene therapy studies, principally as a result of dysregulated host gene expression in the proximity of defined integration sites. Here, we report that cells with characteristics of pluripotent stem cells can be produced from normal human fibroblasts in the absence of reprogramming transcription factors (TFs) during lentiviral (LV) vector–mediated gene transfer. This occurred via induced alterations in host gene and microRNA (miRNA) expression and detrimental changes in karyotype. These findings demonstrate that vector-induced genotoxicity may alone play a role in somatic cell reprogramming derivation and urges caution when using integrating vectors in this setting. Clearer understanding of this process may additionally reveal novel insights into reprogramming pathways.
- Published
- 2010
45. Correction of SCID-X1 using an enhancerless Vav promoter
- Author
-
Elena Almarza, Michael P. Blundell, Steven J. Howe, Juan A. Bueren, Adrian J. Thrasher, Giorgia Santilli, Susannah I. Thornhill, and Fang Zhang
- Subjects
VAV1 ,Genetic enhancement ,Genetic Vectors ,Molecular Sequence Data ,Biology ,X-Linked Combined Immunodeficiency Diseases ,Viral vector ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,Gene Order ,Genetics ,medicine ,Gene silencing ,Animals ,Humans ,Enhancer ,Promoter Regions, Genetic ,Proto-Oncogene Proteins c-vav ,Molecular Biology ,030304 developmental biology ,Common gamma chain ,Regulation of gene expression ,Mice, Knockout ,0303 health sciences ,Severe combined immunodeficiency ,Mice, Inbred BALB C ,Base Sequence ,Lentivirus ,Hematopoietic Stem Cell Transplantation ,Genetic Therapy ,medicine.disease ,Hematopoietic Stem Cells ,3. Good health ,Mice, Inbred C57BL ,Disease Models, Animal ,HEK293 Cells ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,Cancer research ,Molecular Medicine ,Interleukin-2 ,Interleukin Receptor Common gamma Subunit ,Signal Transduction - Abstract
The efficacy of gene therapy for the treatment of inherited immunodeficiency has been highlighted in recent clinical trials, although in some cases complicated by insertional mutagenesis and silencing of vector genomes through methylation. To minimize these effects, we have evaluated the use of regulatory elements that confer reliability of gene expression, but also lack potent indiscriminate enhancer activity. The Vav1 proximal promoter is particularly attractive in this regard and may be useful in situations where high-level or complex regulation of gene expression is not necessary. X-linked severe combined immunodeficiency (SCID-X1) is a good candidate for such an approach, particularly as there may be additional disease-related intrinsic risks of leukemogenesis, and where safety is therefore a paramount concern. We have tested whether lentiviral vectors expressing the common cytokine receptor gamma chain under the control of the proximal Vav1 gene promoter are effective for correction of signaling defects and the disease phenotype. Despite low-level gene expression, we observed near-complete restoration of cytokine-mediated STAT5 phosphorylation in a model cell line. Furthermore, at low vector copy number, highly effective T- and B-lymphocyte reconstitution was achieved in vivo in a murine model of SCID-X1, in both primary and secondary graft recipients. This vector configuration deserves further evaluation and consideration for future clinical trials.
- Published
- 2010
46. A Ubiquitous Chromatin Opening Element (UCOE) Confers Resistance to DNA Methylation–mediated Silencing of Lentiviral Vectors
- Author
-
Fang Zhang, Amy R Frost, Michael Antoniou, Michael P. Blundell, Adrian J. Thrasher, and Olivia Bales
- Subjects
Chromosomal Proteins, Non-Histone ,Genetic Vectors ,Cell Line ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Epigenetics of physical exercise ,Cell Line, Tumor ,Drug Discovery ,Heterogeneous-Nuclear Ribonucleoprotein Group A-B ,Genetics ,Gene silencing ,Animals ,Humans ,Epigenetics ,Promoter Regions, Genetic ,Molecular Biology ,030304 developmental biology ,Regulation of gene expression ,Pharmacology ,0303 health sciences ,biology ,Lentivirus ,Methylation ,DNA Methylation ,Molecular biology ,Chromatin ,Mice, Inbred C57BL ,Histone ,030220 oncology & carcinogenesis ,DNA methylation ,biology.protein ,Molecular Medicine ,Original Article - Abstract
DNA methylation may restrict the activity of gene transfer vectors due to inadvertent silencing. In P19 embryonic carcinoma cells in vitro, we found that transgene expression regulated by the SFFV LTR and EF1 alpha promoter declined rapidly within 16 days, but for A2UCOE derived from the human HNRPA2B1-CBX3 housekeeping gene locus, remained completely stable. Silencing correlated with extensive epigenetic methylation of CpG sites, whereas the A2UCOE was almost completely resistant. Linking of the A2UCOE upstream of the SFFV LTR protected this element from both DNA methylation and silencing. Analysis of engrafted hematopoietic cells in vivo transduced with the same vectors revealed a similar pattern. The A2UCOE displayed little or no methylation in either primary or secondary graft recipients, and gene expression profiles were highly conserved between the two groups. These studies provide convincing evidence that DNA methylation plays a direct role in regulating self-inactivating (SIN) lentiviral transgene expression, and that the stability of expression from the A2UCOE is, at least in part, due to methylation resistance. The A2UCOE therefore has considerable utility for gene therapy applications where reliable and sustained gene expression is desirable.
- Published
- 2010
47. Nonintegrating lentivector vaccines stimulate prolonged T-cell and antibody responses and are effective in tumor therapy
- Author
-
L Apolonia, Gerben Bouma, Mary Collins, David Escors, Michael P. Blundell, Sayandip Mukherjee, Adrian J. Thrasher, and Katarzyna Karwacz
- Subjects
Cellular immunity ,Antibodies, Neoplasm ,T cell ,T-Lymphocytes ,Virus Integration ,Immunology ,Antigen presentation ,Genetic Vectors ,Mice, Transgenic ,Antibodies, Viral ,Microbiology ,Cancer Vaccines ,Viral vector ,Mice ,Immune system ,Antigen ,Virology ,Neoplasms ,Vaccines and Antiviral Agents ,medicine ,Animals ,Hepatitis B Vaccines ,Mice, Inbred BALB C ,biology ,Lentivirus ,medicine.anatomical_structure ,Insect Science ,Humoral immunity ,biology.protein ,Antibody - Abstract
Lentiviral vectors (lentivectors) are effective for stimulation of cell-mediated and humoral immunity following subcutaneous and intramuscular immunization. However, lentivector genome integration carries a risk of perturbation of host gene expression. Here, we demonstrate that lentivectors with multiple mutations that prevent integration are also effective immunogens. First, systemic CD8 + T-cell responses to the model antigen ovalbumin were detected following subcutaneous injection of nonintegrating lentivectors. Transfer of transgenic OT1 T cells demonstrated that antigen presentation persisted for at least 30 days. Furthermore, an enhanced CD8 + T-cell response, peaking at 7 days, was stimulated by coexpression of p38 MAP kinase or an NF-κB activator from the same vector. Second, we demonstrated systemic CD8 + T-cell and antibody responses to the secreted hepatitis B virus (HBV) surface antigen expressed from a nonintegrating lentivector injected intramuscularly. The induction, specificity, and kinetics of antibody production closely mimicked those of natural HBV infection. In this case, both the vector genome and the immune response were maintained for at least 2 months. Together, our data indicate that nonintegrating lentivectors can be employed to generate effective vaccines.
- Published
- 2009
48. Improvement of migratory defects in a murine model of Wiskott-Aldrich syndrome gene therapy
- Author
-
Yolanda Calle, Gerben Bouma, Michael P. Blundell, Adrian J. Thrasher, Christine Kinnon, and Gareth E. Jones
- Subjects
Podosome ,Cell Transplantation ,Genetic enhancement ,Transgene ,Cell ,Green Fluorescent Proteins ,Biology ,Models, Biological ,Viral vector ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Movement ,Drug Discovery ,Genetics ,medicine ,Animals ,Molecular Biology ,030304 developmental biology ,Pharmacology ,Mice, Knockout ,0303 health sciences ,Chemotaxis ,Lentivirus ,Hematopoietic stem cell ,Cell migration ,Dendritic cell ,Dendritic Cells ,Genetic Therapy ,Wiskott-Aldrich Syndrome ,medicine.anatomical_structure ,Langerhans Cells ,Immunology ,Cancer research ,Molecular Medicine ,Dinitrofluorobenzene ,Spleen ,030215 immunology - Abstract
Wiskott–Aldrich syndrome (WAS) is an X-linked hematological disease characterized by immunodeficiency, eczema, and thrombocytopaenia, and shows promise for treatment with hematopoietic stem cell gene therapy. The immunopathology of WAS is attributable at least in part to defects of cell migration and localization as a result of chemotactic, adhesive, and chemokinetic defects. Whereas previous studies using either gammaretroviral or lentiviral vectors have demonstrated variable correction of T-cell proliferation and dendritic cell (DC) cytoarchitecture, we have used a lentiviral vector expressing an eGFP–WASp fusion protein to test the potential for restoration of cell migratory defects. Multilineage expression of the fusion transgene was present for up to 10 months after primary engraftment, and also in secondary recipients analyzed after a further 9 months. Transduced bone marrow–derived dendritic cells (BMDCs) demonstrated recovery of podosome numbers and turnover, while B cells, BMDCs, and Langerhans cells (LCs) exhibited enhanced chemotactic responses to specific stimuli. As an indication of functionality in vivo, splenic marginal zone B cells and a cutaneous contact hypersensitivity (CHS) response to dinitrofluorobenzene (DNFB) were both partially restored. These proof of principle experiments demonstrate that WAS protein (WASp) transgene expression can be successfully maintained long term in primary and secondary recipients, and that it is associated with a significant repair of migratory defects.
- Published
- 2008
49. Cell-specific and efficient expression in mouse and human B cells by a novel hybrid immunoglobulin promoter in a lentiviral vector
- Author
-
Ulrica Brunsberg, H. E. Baxendale, Joanna Sinclair, Kenth Gustafsson, Adrian J. Thrasher, Michael P. Blundell, Steven J. Howe, Rikard Holmdahl, K. L. Laurie, and Waseem Qasim
- Subjects
Male ,Genetic enhancement ,CD3 ,T cell ,Genetic Vectors ,Green Fluorescent Proteins ,Gene Expression ,Immunoglobulins ,CD19 ,Flow cytometry ,Viral vector ,Cell Line ,Mice ,Transduction, Genetic ,Genetics ,medicine ,Animals ,Humans ,Transgenes ,Promoter Regions, Genetic ,Molecular Biology ,Severe combined immunodeficiency ,B-Lymphocytes ,biology ,medicine.diagnostic_test ,Reverse Transcriptase Polymerase Chain Reaction ,Genetic Therapy ,medicine.disease ,Flow Cytometry ,Molecular biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Cell culture ,biology.protein ,HIV-1 ,Molecular Medicine ,Female ,Genetic Engineering - Abstract
The expression of genes specifically in B cells is of great interest in both experimental immunology as well as in future clinical gene therapy. We have constructed a novel enhanced B cell-specific promoter (Igk- E) consisting of an immunoglobulin kappa (Igk) minimal promoter combined with an intronic enhancer sequence and a 30 enhancer sequence from Ig genes. The Igk- E promoter was cloned into a lentiviral vector and used to control expression of enhanced green fluorescent protein (eGFP). Transduction of murine B-cell lymphoma cell lines and activated primary splenic B cells, with IgK-E-eGFP lentivirus, resulted in expression of eGFP, as analysed by flow cytometry, whereas expression in non-B cells was absent. The specificity of the promoter was further examined by transducing Lin bone marrow with Igk-E-eGFP lentivirus and reconstituting lethally irradiated mice. After 16 weeks flow cytometry of lymphoid tissues revealed eGFP expression by CD19(+) cells, but not by CD3(+), CD11b(+), CD11c(+) or Gr-1(+) cells. CD19(+) cells were comprised of both marginal zone B cells and recirculating follicular B cells. Activated human peripheral mononuclear cells were also transduced with Igk-E-eGFP lentivirus under conditions of selective B-cell activation. The Igk-E promoter was able to drive expression of eGFP only in CD19(+) cells, while eGFP was expressed by both spleen focus forming virus and cytomegalovirus constitutive promoters in CD19(+) and CD3(+) lymphocytes. These data demonstrate that in these conditions the Igk-E promoter is cell specific and controls efficient expression of a reporter protein in mouse and human B cells in the context of a lentiviral vector.
- Published
- 2007
50. Lentiviral vectors targeting WASp expression to hematopoietic cells, efficiently transduce and correct cells from WAS patients
- Author
-
Adrian J. Thrasher, Sabine Charrier, R Eckenberg, Loïc Dupré, Alessandro Aiuti, M. Grazia Roncarolo, Federica Cattaneo, Samantha Scaramuzza, Michael P. Blundell, L Jeanson-Leh, Olivier Danos, Anne Galy, Généthon, Immunologie moléculaire et biothérapies innovantes (IMBI), École Pratique des Hautes Études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université d'Évry-Val-d'Essonne (UEVE)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Généthon, Charrier, S, Dupre, L, Scaramuzza, S, JEANSON LEH, L, Blundell, Mp, Danos, O, Cattaneo, F, Aiuti, Alessandro, Eckenberg, R, Thrasher, Aj, Roncarolo, MARIA GRAZIA, and Galy, A.
- Subjects
Transgene ,Genetic enhancement ,T-Lymphocytes ,Blotting, Western ,Genetic Vectors ,Molecular Sequence Data ,Gene Expression ,Antigens, CD34 ,Biology ,Cell Line ,Transduction (genetics) ,Transduction, Genetic ,Genetics ,Humans ,Promoter Regions, Genetic ,Molecular Biology ,Cells, Cultured ,Cell Proliferation ,Regulation of gene expression ,B-Lymphocytes ,Base Sequence ,Lentivirus ,Gene targeting ,Dendritic cell ,Dendritic Cells ,Genetic Therapy ,Sequence Analysis, DNA ,Hematopoietic Stem Cells ,Virology ,Cell biology ,Wiskott-Aldrich Syndrome ,Haematopoiesis ,Microscopy, Fluorescence ,Gene Targeting ,Molecular Medicine ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,Interleukin-2 ,Stem cell ,Wiskott-Aldrich Syndrome Protein - Abstract
Gene therapy has been proposed as a potential treatment for Wiskott-Aldrich syndrome (WAS), a severe primary immune deficiency characterized by multiple hematopoietic-specific cellular defects. In order to develop an optimal lentiviral gene transfer cassette for this application, we compared the performance of several internal promoters in a variety of cell lineages from human WAS patients. Vectors using endogenous promoters derived from short (0.5 kb) or long (1.6 kb) 50 flanking sequences of the WAS gene, expressed the transgene in T, B, dendritic cells as well as CD34(+) progenitor cells, but functioned poorly in non-hematopoietic cells. Defects of T-cell proliferation and interleukin-2 production, and the cytoskeletal anomalies in WAS dendritic cells were also corrected. The levels of reconstitution were comparable to those obtained following transduction with similar lentiviral vectors incorporating constitutive PGK-1, EF1-alpha promoters or the spleen focus forming virus gammaretroviral LTR. Thus, native regulatory sequences target the expression of the therapeutic WAS transgene to the hematopoietic system, as is naturally the case for WAS, and are effective for correction of multiple cellular defects. These vectors may have significant advantages for clinical application in terms of natural gene regulation, and reduction in the potential for adverse mutagenic events. Gene therapy has been proposed as a potential treatment for Wiskott–Aldrich syndrome (WAS), a severe primary immune deficiency characterized by multiple hematopoietic-specific cellular defects. In order to develop an optimal lentiviral gene transfer cassette for this application, we compared the performance of several internal promoters in a variety of cell lineages from human WAS patients. Vectors using endogenous promoters derived from short (0.5 kb) or long (1.6 kb) 5' flanking sequences of the WAS gene, expressed the transgene in T, B, dendritic cells as well as CD34+ progenitor cells, but functioned poorly in non-hematopoietic cells. Defects of T-cell proliferation and interleukin-2 production, and the cytoskeletal anomalies in WAS dendritic cells were also corrected. The levels of reconstitution were comparable to those obtained following transduction with similar lentiviral vectors incorporating constitutive PGK-1, EF1-alpha promoters or the spleen focus forming virus gammaretroviral LTR. Thus, native regulatory sequences target the expression of the therapeutic WAS transgene to the hematopoietic system, as is naturally the case for WAS, and are effective for correction of multiple cellular defects. These vectors may have significant advantages for clinical application in terms of natural gene regulation, and reduction in the potential for adverse mutagenic events
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.