Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Methods Mol Biol ; 2261: 213-227, 2021.
Article in English | MEDLINE | ID: mdl-33420992

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.


Subject(s)
Antigens/analysis , Flow Cytometry , Immunophenotyping , Proteins/analysis , Proteomics , Fluorescent Dyes/chemistry , Humans
3.
Sci Rep ; 9(1): 11592, 2019 08 12.
Article in English | MEDLINE | ID: mdl-31406195

ABSTRACT

In utero gene therapy (IUGT) to the fetal hematopoietic compartment could be used to treat congenital blood disorders such as ß-thalassemia. A humanised mouse model of ß-thalassemia was used, in which heterozygous animals are anaemic with splenomegaly and extramedullary hematopoiesis. Intrahepatic in utero injections of a ß globin-expressing lentiviral vector (GLOBE), were performed in fetuses at E13.5 of gestation. We analysed animals at 12 and 32 weeks of age, for vector copy number in bone marrow, peripheral blood liver and spleen and we performed integration site analysis. Compared to noninjected heterozygous animals IUGT normalised blood haemoglobin levels and spleen weight. Integration site analysis showed polyclonality. The left ventricular ejection fraction measured using magnetic resonance imaging (MRI) in treated heterozygous animals was similar to that of normal non-ß-thalassemic mice but significantly higher than untreated heterozygous thalassemia mice suggesting that IUGT ameliorated poor cardiac function. GLOBE LV-mediated IUGT normalised the haematological and anatomical phenotype in a heterozygous humanised model of ß-thalassemia.


Subject(s)
Genetic Therapy , Heterozygote , Magnetic Resonance Imaging/methods , Animals , Female , Humans , Mice , Phenotype , Pregnancy , beta-Thalassemia/genetics
4.
Stem Cells Dev ; 27(8): 515-523, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29482456

ABSTRACT

Clinical success of in utero transplantation (IUT) using allogeneic hematopoietic stem cells (HSCs) has been limited to fetuses that lack an immune response to allogeneic cells due to severe immunological defects, and where transplanted genetically normal cells have a proliferative or survival advantage. Amniotic fluid (AF) is an autologous source of stem cells with hematopoietic potential that could be used to treat congenital blood disorders. We compared the ability of congenic and allogeneic mouse AF stem cells (AFSC) to engraft the hematopoietic system of time-mated C57BL/6J mice (E13.5). At 4 and 16 weeks of age, multilineage donor engraftment was higher in congenic versus allogeneic animals. In vitro mixed lymphocyte reaction confirmed an immune response in the allogeneic group with higher CD4 and CD8 cell counts and increased proliferation of stimulated lymphocytes. IUT with congenic cells resulted in 100% of donor animals having chimerism of around 8% and successful hematopoietic long-term engraftment in immune-competent mice when compared with IUT with allogeneic cells. AFSCs may be useful for autologous cell/gene therapy approaches in fetuses diagnosed with congenital hematopoietic disorders.


Subject(s)
Graft Survival , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/immunology , Immunocompetence , Amniotic Fluid/cytology , Amniotic Fluid/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Female , Fetus , Hematopoietic Stem Cells/cytology , Injections, Intraperitoneal , Lymphocyte Count , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred C57BL , Transplantation Chimera , Transplantation, Homologous , Transplantation, Isogeneic , Uterus/immunology
5.
J Allergy Clin Immunol ; 142(3): 904-913.e3, 2018 09.
Article in English | MEDLINE | ID: mdl-29355678

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.


Subject(s)
CD8-Positive T-Lymphocytes/transplantation , Lymphocytic Choriomeningitis/therapy , Lymphohistiocytosis, Hemophagocytic/therapy , Perforin/genetics , Animals , Cell Line, Tumor , Child, Preschool , Genetic Therapy , Humans , Lymphocytic choriomeningitis virus , Male , Mice, Inbred C57BL , Mice, Knockout
6.
J Exp Med ; 214(1): 59-71, 2017 01.
Article in English | MEDLINE | ID: mdl-27994071

ABSTRACT

The importance of actin dynamics in the activation of the inflammasome is becoming increasingly apparent. IL-1ß, 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-1ß, and their cells also secreted more IL-18 but not IL-1ß 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.


Subject(s)
Hereditary Autoinflammatory Diseases/genetics , Immunologic Deficiency Syndromes/genetics , Microfilament Proteins/genetics , Mutation, Missense , Thrombocytopenia/genetics , Actins/metabolism , Child , Female , Hereditary Autoinflammatory Diseases/etiology , Humans , Immunologic Deficiency Syndromes/etiology , Inflammasomes/physiology , Interleukin-18/blood , Microfilament Proteins/physiology , Phagocytosis , Thrombocytopenia/etiology
7.
J Leukoc Biol ; 99(5): 699-710, 2016 05.
Article in English | MEDLINE | ID: mdl-26590149

ABSTRACT

The immunological synapse is a highly structured and molecularly dynamic interface between communicating immune cells. Although the immunological synapse promotes T cell activation by dendritic cells, the specific organization of the immunological synapse on the dendritic cell side in response to T cell engagement is largely unknown. In this study, confocal and electron microscopy techniques were used to investigate the role of dendritic cell actin regulation in immunological synapse formation, stabilization, and function. In the dendritic cell-restricted absence of the Wiskott-Aldrich syndrome protein, an important regulator of the actin cytoskeleton in hematopoietic cells, the immunological synapse contact with T cells occupied a significantly reduced surface area. At a molecular level, the actin network localized to the immunological synapse exhibited reduced stability, in particular, of the actin-related protein-2/3-dependent, short-filament network. This was associated with decreased polarization of dendritic cell-associated ICAM-1 and MHC class II, which was partially dependent on Wiskott-Aldrich syndrome protein phosphorylation. With the use of supported planar lipid bilayers incorporating anti-ICAM-1 and anti-MHC class II antibodies, the dendritic cell actin cytoskeleton organized into recognizable synaptic structures but interestingly, formed Wiskott-Aldrich syndrome protein-dependent podosomes within this area. These findings demonstrate that intrinsic dendritic cell cytoskeletal remodeling is a key regulatory component of normal immunological synapse formation, likely through consolidation of adhesive interaction and modulation of immunological synapse stability.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Communication/immunology , Dendritic Cells/immunology , Immunological Synapses/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Wiskott-Aldrich Syndrome Protein/metabolism , Animals , Fluorescence Recovery After Photobleaching , Intercellular Adhesion Molecule-1/metabolism , Lipid Bilayers/metabolism , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Podosomes/metabolism
8.
Stem Cells ; 33(1): 122-32, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25186828

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.


Subject(s)
Amniotic Fluid/cytology , Amniotic Fluid/metabolism , Antigens, CD34/biosynthesis , Hematopoietic Stem Cell Transplantation/methods , Animals , Cell- and Tissue-Based Therapy , Female , Fetus/surgery , Mice , Mice, Inbred NOD , Mice, SCID , Pregnancy , Sheep , Transplantation, Autologous , Transplantation, Heterologous
9.
Mol Ther ; 23(4): 737-45, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25523759

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 with >30% 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.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Gene Transfer Techniques , Hematopoietic Stem Cells/metabolism , Killer Cells, Natural/immunology , Lymphohistiocytosis, Hemophagocytic/therapy , Perforin/genetics , Animals , Lymphohistiocytosis, Hemophagocytic/immunology , Mice , Mice, Transgenic , Phenotype
10.
Cell Reprogram ; 16(5): 331-44, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25162836

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.


Subject(s)
Amniotic Fluid/cytology , Cryopreservation , Down Syndrome , Induced Pluripotent Stem Cells/cytology , Models, Biological , Animals , Female , Humans , Mice , Pregnancy , Prenatal Diagnosis
11.
Int J Biochem Cell Biol ; 50: 47-54, 2014 May.
Article in English | MEDLINE | ID: mdl-24508783

ABSTRACT

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-a 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.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/enzymology , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Membrane Structures/enzymology , Cell Movement/physiology , Dendritic Cells/metabolism , Extracellular Matrix/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Transfection , Wiskott-Aldrich Syndrome Protein/metabolism
12.
Mol Ther ; 22(3): 607-622, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24256635

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.


Subject(s)
Adenosine Deaminase/deficiency , Adenosine Deaminase/genetics , Agammaglobulinemia/immunology , Agammaglobulinemia/therapy , Genetic Vectors/adverse effects , Lentivirus/genetics , Peptide Elongation Factor 1/genetics , Severe Combined Immunodeficiency/immunology , Severe Combined Immunodeficiency/therapy , Adenosine Deaminase/immunology , Adenosine Deaminase/metabolism , Agammaglobulinemia/genetics , Agammaglobulinemia/pathology , Animals , B-Lymphocytes/immunology , Cells, Cultured , Disease Models, Animal , Female , HEK293 Cells , HT29 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/pathology , T-Lymphocytes/immunology , Transduction, Genetic , Virus Integration
13.
J Invest Dermatol ; 133(5): 1221-30, 2013 May.
Article in English | MEDLINE | ID: mdl-23303453

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.


Subject(s)
Alopecia/physiopathology , CD4-Positive T-Lymphocytes/pathology , Cell Proliferation , Hair Color/physiology , Pigmentation/physiology , Thymus Gland/transplantation , Transplantation, Heterologous , Adoptive Transfer , Alopecia/pathology , Animals , CD4-Positive T-Lymphocytes/physiology , Disease Models, Animal , Hair Follicle/pathology , Hair Follicle/physiology , Homeodomain Proteins/genetics , Humans , In Vitro Techniques , Melanocytes/pathology , Melanocytes/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Up-Regulation/physiology
14.
Blood ; 121(13): 2542-52, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23305739

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.


Subject(s)
Basement Membrane/physiology , Cell Surface Extensions/physiology , Extracellular Matrix/metabolism , Megakaryocytes/physiology , Animals , Basement Membrane/metabolism , Blood Platelets/metabolism , Blood Platelets/physiology , Cell Surface Extensions/metabolism , Cells, Cultured , Fibrinogen/metabolism , HEK293 Cells , Humans , Infant, Newborn , Matrix Metalloproteinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myosin Type II/metabolism
15.
Stem Cells Dev ; 22(3): 444-58, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23050522

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.


Subject(s)
Amniotic Fluid/cytology , Antigens, Differentiation/metabolism , Histone Deacetylase Inhibitors/pharmacology , Pluripotent Stem Cells/metabolism , Valproic Acid/pharmacology , Animals , Antigens, Differentiation/genetics , Cell Proliferation , Cell Shape , Cells, Cultured , Culture Media , Embryonic Stem Cells/metabolism , Female , Gene Expression , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Neoplasms, Experimental/pathology , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/physiology , Pluripotent Stem Cells/transplantation , Pregnancy , Pregnancy Trimester, Second , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Telomere/metabolism , Teratoma/pathology , Up-Regulation/drug effects
16.
Br J Haematol ; 158(6): 778-87, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22816563

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.


Subject(s)
Bone Marrow/pathology , Cell Adhesion/physiology , Colony-Stimulating Factors/pharmacology , Graft Survival/physiology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/pathology , Stem Cell Niche/physiology , Adult , Animals , Antigens, CD34/analysis , Cells, Cultured/drug effects , Cells, Cultured/transplantation , Hematopoietic Stem Cells/drug effects , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Rats , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology , Specific Pathogen-Free Organisms
17.
Mol Ther ; 20(10): 1953-67, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22760542

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.


Subject(s)
Amniotic Fluid/drug effects , Histone Deacetylase Inhibitors/pharmacology , Induced Pluripotent Stem Cells/drug effects , Valproic Acid/pharmacology , Amniotic Fluid/cytology , Cell Differentiation , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Genome, Human , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Karyotyping , Kinetics , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Phenotype , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Sequence Analysis, DNA , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome , Transgenes , X Chromosome Inactivation/drug effects
18.
Mol Ther ; 20(7): 1400-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22434141

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 ß-globin gene (ß-LCR). We show that the ß-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 ß-LCR did not alter the mutagenic potential of the vector in in vitro mutagenesis (IM) assays although microarray analysis showed that the ß-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.


Subject(s)
Erythroid Precursor Cells/metabolism , Locus Control Region , Peptide Elongation Factor 1/genetics , Peptide Elongation Factors/genetics , beta-Globins/genetics , Adenosine Deaminase/genetics , Animals , Cell Line , Gene Expression Regulation , Genetic Therapy/methods , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , HEK293 Cells , Hematopoietic Stem Cells , Humans , Jurkat Cells , Lentivirus/genetics , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/therapy , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , U937 Cells , Up-Regulation
19.
Haematologica ; 97(5): 687-91, 2012 May.
Article in English | MEDLINE | ID: mdl-22133775

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.


Subject(s)
Actin Cytoskeleton/physiology , Calpain/metabolism , Cell Membrane Structures/metabolism , Tyrosine/metabolism , Wiskott-Aldrich Syndrome Protein/physiology , Animals , Cell Adhesion , Cell Movement , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/metabolism , Fluorescent Antibody Technique , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Myeloid Cells/cytology , Myeloid Cells/metabolism , Phosphorylation , Protein Binding
20.
Blood ; 118(9): 2492-501, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21690559

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.


Subject(s)
Cytoskeleton/ultrastructure , Dendritic Cells/ultrastructure , Immunological Synapses/ultrastructure , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Wiskott-Aldrich Syndrome Protein/physiology , Animals , Bacterial Proteins/analysis , Bacterial Proteins/genetics , Calcium Signaling/immunology , Cell Movement , Crosses, Genetic , Genes, Reporter , Genetic Complementation Test , Humans , Immunological Synapses/immunology , Luminescent Proteins/analysis , Luminescent Proteins/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovalbumin/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/physiology , T-Lymphocytes/ultrastructure , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome/pathology , Wiskott-Aldrich Syndrome Protein/deficiency , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome Protein/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...