Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
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.
Ann Med Surg (Lond) ; 48: 65-68, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31719979

ABSTRACT

BACKGROUND: It is now recognised that the majority of breast surgery can be safely undertaken as day case procedures. We aimed to evaluate the effect of pectoral nerve (Pecs2) blocks on recovery parameters and day case rates in patients undergoing mastectomy for breast cancer. METHODS: A prospective cohort study was performed in a single NHS Foundation trust between 1st April 2014 and 31st December 2016. Visual analogue scale (VAS) pain scores (0-10) at 4 and 8 h, episodes of post-operative nausea ±â€¯vomiting (PONV), opioid use and day case outcome were compared between Pecs2 and no Pecs2 groups. RESULTS: 22 patients underwent general anaesthesia (GA) + Pecs2 block and 30 GA ± local anaesthetic infiltration.Mean pain scores were significantly lower in the Pecs2 (2.5) vs no Pecs2 (4.6) group at 4 h (p = 0.0132) and 8 h, Pecs2 (1.9) vs no Pecs2 (3.6) (p = 0.0038).Episodes of PONV requiring additional anti-emetic were lower and statistically significant in the Pecs2 group (2/22, 9%) than the no Pecs2 group (14/30, 46%), (p = 0.005).Additional opioid use was significantly lower in the Pecs2 group (4/22, 18%) than in the no Pecs2 group (14/30, 46%) (p = 0.0423).18 patients in the Pecs2 group were discharged the same day in contrast to just 3 patients in the no Pecs2 group. This was highly statistically significant (p = 0.0001). CONCLUSIONS: Pecs2 blocks can significantly reduce post-operative pain, nausea and vomiting in patients undergoing mastectomy. Their use can enable units to achieve high day-case mastectomy rates.

4.
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
5.
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
6.
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
7.
Cytotherapy ; 19(6): 710-720, 2017 06.
Article in English | MEDLINE | ID: mdl-28428057

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.


Subject(s)
Antigens, CD34/metabolism , Fetal Blood/cytology , Immunotherapy/methods , Killer Cells, Natural/immunology , Cell Culture Techniques , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Cytotoxicity, Immunologic , Humans , Interleukin-2/pharmacology , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Leukemia, Myeloid, Acute/therapy
8.
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
9.
Sci Rep ; 6: 22097, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26915707

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.


Subject(s)
Cell Proliferation , Hematopoiesis , Killer Cells, Natural/physiology , T-Lymphocytes, Regulatory/physiology , Adoptive Transfer , Animals , Humans , Killer Cells, Natural/immunology , Mice , T-Lymphocytes, Regulatory/immunology
10.
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
11.
PLoS One ; 10(10): e0138623, 2015.
Article in English | MEDLINE | ID: mdl-26465138

ABSTRACT

Cord blood (CB) is increasingly used as a source of hematopoietic stem cells (HSC) for transplantation. Low incidence and severity of graft-versus-host disease (GvHD) and a robust graft-versus-leukemia (GvL) effect are observed following CB transplantation (CBT). However, its main disadvantages are a limited number of HSC per unit, delayed immune reconstitution and a higher incidence of infection. Unmanipulated grafts contain accessory cells that may facilitate HSC engraftment. Therefore, the effects of accessory cells, particularly natural killer (NK) cells, on human CB HSC (CBSC) functions were assessed in vitro and in vivo. CBSC cultured with autologous CB NK cells showed higher levels of CXCR4 expression, a higher migration index and a higher number of colony forming units (CFU) after short-term and long-term cultures. We found that CBSC secreted CXCL9 following interaction with CB NK cells. In addition, recombinant CXCL9 increased CBSC clonogenicity, recapitulating the effect observed of CB NK cells on CBSC. Moreover, the co-infusion of CBSC with CB NK cells led to a higher level of CBSC engraftment in NSG mouse model. The results presented in this work offer the basis for an alternative approach to enhance HSC engraftment that could improve the outcome of CBT.


Subject(s)
Cord Blood Stem Cell Transplantation/methods , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/cytology , Killer Cells, Natural/cytology , Stem Cells/cytology , Animals , Cell Movement , Chemokine CXCL9/metabolism , Cytokines/metabolism , Female , Fetal Blood/cytology , Gene Expression Regulation , Graft vs Host Disease/physiopathology , Graft vs Leukemia Effect , Humans , Interleukin-15/metabolism , Leukocytes, Mononuclear/cytology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Oligonucleotide Array Sequence Analysis , Recombinant Proteins/metabolism
12.
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
13.
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
14.
Blood ; 124(16): 2514-22, 2014 Oct 16.
Article in English | MEDLINE | ID: mdl-25185261

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.


Subject(s)
Epstein-Barr Virus Infections/complications , Genetic Therapy , Immunotherapy, Adoptive , Lymphoma/therapy , Lymphoma/virology , T-Lymphocytes, Cytotoxic/transplantation , T-Lymphocytes, Cytotoxic/virology , Animals , Calcineurin/genetics , Calcineurin Inhibitors/pharmacology , Drug Resistance , Genetic Engineering/methods , Genetic Therapy/methods , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/isolation & purification , Humans , Immunosuppressive Agents/pharmacology , Immunotherapy, Adoptive/methods , Lymphoma/genetics , Lymphoma/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Mutation , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/metabolism , Tacrolimus/pharmacology , Transduction, Genetic
15.
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
16.
PLoS One ; 9(1): e87086, 2014.
Article in English | MEDLINE | ID: mdl-24489840

ABSTRACT

Adoptive natural killer (NK) cell therapy relies on the acquisition of large numbers of NK cells that are cytotoxic but not exhausted. NK cell differentiation from hematopoietic stem cells (HSC) has become an alluring option for NK cell therapy, with umbilical cord blood (UCB) and mobilized peripheral blood (PBCD34(+)) being the most accessible HSC sources as collection procedures are less invasive. In this study we compared the capacity of frozen or freshly isolated UCB hematopoietic stem cells (CBCD34(+)) and frozen PBCD34(+) to generate NK cells in vitro. By modifying a previously published protocol, we showed that frozen CBCD34(+) cultures generated higher NK cell numbers without loss of function compared to fresh CBCD34(+) cultures. NK cells generated from CBCD34(+) and PBCD34(+) expressed low levels of killer-cell immunoglobulin-like receptors but high levels of activating receptors and of the myeloid marker CD33. However, blocking studies showed that CD33 expression did not impact on the functions of the generated cells. CBCD34(+)-NK cells exhibited increased capacity to secrete IFN-γ and kill K562 in vitro and in vivo as compared to PBCD34(+)-NK cells. Moreover, K562 killing by the generated NK cells could be further enhanced by IL-12 stimulation. Our data indicate that the use of frozen CBCD34(+) for the production of NK cells in vitro results in higher cell numbers than PBCD34(+), without jeopardizing their functionality, rendering them suitable for NK cell immunotherapy. The results presented here provide an optimal strategy to generate NK cells in vitro for immunotherapy that exhibit enhanced effector function when compared to alternate sources of HSC.


Subject(s)
Cytotoxicity, Immunologic , Fetal Blood/cytology , Hematopoietic Stem Cells/cytology , Killer Cells, Natural/cytology , Adoptive Transfer , Animals , Antigens, CD34/genetics , Antigens, CD34/immunology , Biomarkers/metabolism , Cell Differentiation , Cells, Cultured , Coculture Techniques , Cryopreservation , Fetal Blood/immunology , Gene Expression , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/immunology , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/metabolism , Interleukin-12/pharmacology , K562 Cells , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/transplantation , Mice , Mice, Inbred NOD , Sialic Acid Binding Ig-like Lectin 3/genetics , Sialic Acid Binding Ig-like Lectin 3/immunology
17.
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
18.
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
19.
Haematologica ; 98(8): 1300-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23445877

ABSTRACT

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.


Subject(s)
Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/metabolism , Wiskott-Aldrich Syndrome Protein/deficiency , Wiskott-Aldrich Syndrome/metabolism , Wiskott-Aldrich Syndrome/therapy , Animals , Hematopoietic Stem Cells/drug effects , Humans , Male , Mice , Mice, 129 Strain , Mice, Knockout , Recombinant Proteins/pharmacology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...