Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
2.
J Autoimmun ; 138: 103051, 2023 07.
Article in English | MEDLINE | ID: mdl-37224733

ABSTRACT

Tolerogenic dendritic cells play a critical role in promoting antigen-specific tolerance via dampening of T cell responses, induction of pathogenic T cell exhaustion and antigen-specific regulatory T cells. Here we efficiently generate tolerogenic dendritic cells by genetic engineering of monocytes with lentiviral vectors co-encoding for immunodominant antigen-derived peptides and IL-10. These transduced dendritic cells (designated DCIL-10/Ag) secrete IL-10 and efficiently downregulate antigen-specific CD4+ and CD8+ T cell responses from healthy subjects and celiac disease patients in vitro. In addition, DCIL-10/Ag induce antigen-specific CD49b+LAG-3+ T cells, which display the T regulatory type 1 (Tr1) cell gene signature. Administration of DCIL-10/Ag resulted in the induction of antigen-specific Tr1 cells in chimeric transplanted mice and the prevention of type 1 diabetes in pre-clinical disease models. Subsequent transfer of these antigen-specific T cells completely prevented type 1 diabetes development. Collectively these data indicate that DCIL-10/Ag represent a platform to induce stable antigen-specific tolerance to control T-cell mediated diseases.


Subject(s)
Diabetes Mellitus, Type 1 , Interleukin-10 , Animals , Mice , Antigens , Dendritic Cells/metabolism , Diabetes Mellitus, Type 1/therapy , Diabetes Mellitus, Type 1/metabolism , Immune Tolerance , Interleukin-10/genetics , Interleukin-10/metabolism , T-Lymphocytes, Regulatory/metabolism , Humans , Celiac Disease
3.
Sci Transl Med ; 15(698): eade3856, 2023 05 31.
Article in English | MEDLINE | ID: mdl-37256935

ABSTRACT

Dysregulation of the interleukin-1 (IL-1) pathway leads to immune diseases that can result in chronic tissue and organ inflammation. Although IL-1 blockade has shown promise in ameliorating these symptoms and improving patients' quality of life, there is an urgent need for more effective, long-lasting treatments. We developed a lentivirus (LV)-mediated gene transfer strategy using transplanted autologous hematopoietic stem/progenitor cells (HSPCs) as a source of IL-1 receptor antagonist (IL-1RA) for systemic delivery to tissues and organs. Transplantation of mouse and human HSPCs transduced with an IL-1RA-encoding LV ensured stable IL-1RA production while maintaining the clonogenic and differentiation capacities of HSPCs in vivo. We examined the efficacy of cell-mediated IL-1RA delivery in three models of IL-1-dependent inflammation, for which treatment hindered neutrophil recruitment in an inducible model of gout, prevented systemic and multi-tissue inflammation in a genetic model of cryopyrin-associated periodic syndromes, and reduced disease severity in an experimental autoimmune encephalomyelitis model of multiple sclerosis. Our findings demonstrate HSPC-mediated IL-1RA delivery as a potential therapeutic modality that can be exploited to suppress tissue and organ inflammation in diverse immune-related diseases involving IL-1-driven inflammation.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Interleukin 1 Receptor Antagonist Protein , Animals , Humans , Encephalomyelitis, Autoimmune, Experimental/therapy , Inflammation/therapy , Interleukin-1 , Lentivirus , Quality of Life , Mice
4.
Cell Rep ; 42(3): 112193, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36870061

ABSTRACT

Interleukin (IL)-10 is a main player in peripheral immune tolerance, the physiological mechanism preventing immune reactions to self/harmless antigens. Here, we investigate IL-10-induced molecular mechanisms generating tolerogenic dendritic cells (tolDC) from monocytes. Using genomic studies, we show that IL-10 induces a pattern of accessible enhancers exploited by aryl hydrocarbon receptor (AHR) to promote expression of a set of core genes. We demonstrate that AHR activity occurs downstream of IL-10 signaling in myeloid cells and is required for the induction of tolerogenic activities in DC. Analyses of circulating DCs show that IL-10/AHR genomic signature is active in vivo in health. In multiple sclerosis patients, we instead observe significantly altered signature correlating with functional defects and reduced frequencies of IL-10-induced-tolDC in vitro and in vivo. Our studies identify molecular mechanisms controlling tolerogenic activities in human myeloid cells and may help in designing therapies to re-establish immune tolerance.


Subject(s)
Interleukin-10 , Receptors, Aryl Hydrocarbon , Humans , Dendritic Cells/metabolism , Immune Tolerance , Interleukin-10/metabolism , Monocytes/metabolism , Receptors, Aryl Hydrocarbon/genetics , Receptors, Aryl Hydrocarbon/metabolism
6.
Haematologica ; 106(10): 2588-2597, 2021 10 01.
Article in English | MEDLINE | ID: mdl-33054128

ABSTRACT

Type 1 regulatory (Tr1) T cells induced by enforced expression of IL-10 (LV-10) are being developed as a novel treatment for chemotherapy-resistant myeloid leukemias. In vivo, LV-10 cells do not cause graft vs host disease while mediating graft vs leukemia (GvL) effect against adult acute myeloid leukemia (AML). Since pediatric AML (pAML) and adult AML are different on a genetic and epigenetic level, we investigate herein whether LV-10 cells also efficiently kill pAML cells. We show that the majority of primary pAML are killed by LV-10 cells, with different levels of sensitivity to killing. Transcriptionally, pAML sensitive to LV-10 killing expressed a myeloid maturation signature. Overlaying the signatures of sensitive and resistant pAML onto the public NCI TARGET pAML dataset revealed that sensitive pAML clustered with M5 monocytic pAML and pAML with MLL rearrangement. Resistant pAML clustered with myelomonocytic leukemias and those bearing the core binding factor translocations inv(16) or t(8;21)(RUNX1-RUNX1T1). Furthermore, resistant pAML upregulated the membrane glycoprotein CD200, which binds to the inhibitory receptor CD200R1 on LV-10 cells. To examine if CD200 expression on target cells can impair LV-10 cell function, we overexpressed CD200 in myeloid leukemia cell lines ordinarily sensitive to LV-10 killing. Indeed, LV-10 cells degranulated less and killed fewer CD200-overexpressing cells compared to controls, indicating that pAML can utilize CD200 expression for immune evasion. Altogether, the majority of pAML are killed by LV-10 cells in vitro, supporting further LV-10 cell development as an innovative cell therapy for pAML.


Subject(s)
Leukemia, Myeloid, Acute , T-Lymphocytes, Regulatory , Adult , CD4-Positive T-Lymphocytes , Child , Graft vs Leukemia Effect , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Translocation, Genetic
7.
Front Immunol ; 11: 1260, 2020.
Article in English | MEDLINE | ID: mdl-32695103

ABSTRACT

The prominent role of dendritic cells (DC) in promoting tolerance and the development of methods to generate clinical grade products allowed the clinical application of tolerogenic DC (tolDC)-based therapies for controlling unwanted immune responses. We established an efficient method to generate tolerogenic human DC, producing supra-physiological levels of IL-10, by genetically engineering monocyte-derived DC with a bidirectional Lentiviral Vector (bdLV) encoding for IL-10 and a marker gene. DCIL-10 are mature DC, modulate T cell responses, promote T regulatory cells, and are phenotypically and functionally stable upon stimulation. Adoptive transfer of human DCIL-10 in a humanized mouse model dampens allogeneic T cell recall responses, while murine DCIL-10 delays acute graft-vs.-host disease in mice. Our report outlines an efficient method to transduce human myeloid cells with large-size LV and shows that stable over-expression of IL-10 generates an effective cell product for future clinical applications in the contest of allogeneic transplantation.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Genetic Vectors/genetics , Immune Tolerance , Interleukin-10/genetics , Lentivirus/genetics , Transduction, Genetic , Animals , Female , Gene Expression , Humans , Immune Tolerance/genetics , Immunophenotyping , Mice , Monocytes/immunology , Monocytes/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
8.
Mol Ther ; 25(10): 2254-2269, 2017 10 04.
Article in English | MEDLINE | ID: mdl-28807569

ABSTRACT

T regulatory cells (Tregs) play a key role in modulating T cell responses. Clinical trials showed that Tregs modulate graft-versus-host disease (GvHD) after allogeneic hematopoietic stem cell transplantation (allo-HSCT). However, their ability to mediate anti-leukemic activity (graft-versus-leukemia [GvL]) is largely unknown. Enforced interleukin-10 (IL-10) expression converts human CD4+ T cells into T regulatory type 1 (Tr1)-like (CD4IL-10) cells that suppress effector T cells in vitro and xenoGvHD in humanized mouse models. In the present study, we show that CD4IL-10 cells mediate anti-leukemic effects in vitro and in vivo in a human leukocyte antigen (HLA) class I-dependent but antigen-independent manner. The cytotoxicity mediated by CD4IL-10 cells is granzyme B (GzB) dependent, is specific for CD13+ target cells, and requires CD54 and CD112 expression on primary leukemic target blasts. CD4IL-10 cells adoptively transferred in humanized mouse models directly mediate anti-tumor and anti-leukemic effects. In addition, when co-transferred with peripheral blood mononuclear cells (PBMCs), CD4IL-10 cells contribute to the GvL activity but suppress xenoGvHD mediated by the PBMCs. These findings provide for the first time a strong rationale for CD4IL-10 cell immunotherapy to prevent GvHD and promote GvL in allo-HSCT for myeloid malignancies.


Subject(s)
Interleukin-10/metabolism , Leukemia, Myeloid/therapy , Leukocytes, Mononuclear/metabolism , T-Lymphocytes, Regulatory/metabolism , CD4-Positive T-Lymphocytes , Humans , Immunotherapy , Leukemia, Myeloid/immunology , Leukemia, Myeloid/metabolism , Models, Biological
9.
Mol Ther ; 20(9): 1778-90, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22692497

ABSTRACT

Type 1 regulatory T (Tr1) cells are an inducible subset of CD4(+) Tr cells characterized by high levels of interleukin (IL)-10 production and regulatory properties. Several protocols to generate human Tr1 cells have been developed in vitro. However, the resulting population includes a significant fraction of contaminating non-Tr1 cells, representing a major bottleneck for clinical application of Tr1 cell therapy. We generated an homogeneous IL-10-producing Tr1 cell population by transducing human CD4(+) T cells with a bidirectional lentiviral vector (LV) encoding for human IL-10 and the marker gene, green fluorescent protein (GFP), which are independently coexpressed. The resulting GFP(+) LV-IL-10-transduced human CD4(+) T (CD4(LV-IL-10)) cells expressed, upon T-cell receptor (TCR) activation, high levels of IL-10 and concomitant low levels of IL-4, and markers associated with IL-10. Moreover, CD4(LV-IL-10) T cells displayed typical Tr1 features: the anergic phenotype, the IL-10, and transforming growth factor (TGF)-ß dependent suppression of allogeneic T-cell responses, and the ability to suppress in a cell-to-cell contact independent manner in vitro. CD4(LV-IL-10) T cells were able to control xeno graft-versus-host disease (GvHD), demonstrating their suppressive function in vivo. These results show that constitutive over-expression of IL-10 in human CD4(+) T cells leads to a stable cell population that recapitulates the phenotype and function of Tr1 cells.


Subject(s)
Gene Expression , Graft vs Host Disease/prevention & control , Immunomodulation , Interleukin-10/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation , Clonal Anergy , Dendritic Cells/cytology , Dendritic Cells/immunology , Female , Genes, Reporter , Genetic Vectors , Graft vs Host Disease/immunology , Green Fluorescent Proteins , Humans , Interleukin-10/biosynthesis , Interleukin-10/genetics , Interleukin-4/immunology , Lentivirus/genetics , Lymphocyte Activation/immunology , Mice , Mice, Nude , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/transplantation , Transduction, Genetic
10.
N Engl J Med ; 360(5): 447-58, 2009 Jan 29.
Article in English | MEDLINE | ID: mdl-19179314

ABSTRACT

BACKGROUND: We investigated the long-term outcome of gene therapy for severe combined immunodeficiency (SCID) due to the lack of adenosine deaminase (ADA), a fatal disorder of purine metabolism and immunodeficiency. METHODS: We infused autologous CD34+ bone marrow cells transduced with a retroviral vector containing the ADA gene into 10 children with SCID due to ADA deficiency who lacked an HLA-identical sibling donor, after nonmyeloablative conditioning with busulfan. Enzyme-replacement therapy was not given after infusion of the cells. RESULTS: All patients are alive after a median follow-up of 4.0 years (range, 1.8 to 8.0). Transduced hematopoietic stem cells have stably engrafted and differentiated into myeloid cells containing ADA (mean range at 1 year in bone marrow lineages, 3.5 to 8.9%) and lymphoid cells (mean range in peripheral blood, 52.4 to 88.0%). Eight patients do not require enzyme-replacement therapy, their blood cells continue to express ADA, and they have no signs of defective detoxification of purine metabolites. Nine patients had immune reconstitution with increases in T-cell counts (median count at 3 years, 1.07x10(9) per liter) and normalization of T-cell function. In the five patients in whom intravenous immune globulin replacement was discontinued, antigen-specific antibody responses were elicited after exposure to vaccines or viral antigens. Effective protection against infections and improvement in physical development made a normal lifestyle possible. Serious adverse events included prolonged neutropenia (in two patients), hypertension (in one), central-venous-catheter-related infections (in two), Epstein-Barr virus reactivation (in one), and autoimmune hepatitis (in one). CONCLUSIONS: Gene therapy, combined with reduced-intensity conditioning, is a safe and effective treatment for SCID in patients with ADA deficiency. (ClinicalTrials.gov numbers, NCT00598481 and NCT00599781.)


Subject(s)
Adenosine Deaminase/genetics , Antigens, CD34/genetics , Genetic Therapy , Hematopoietic Stem Cell Transplantation , Severe Combined Immunodeficiency/therapy , Adenosine Deaminase/deficiency , Bone Marrow Cells/immunology , Child, Preschool , Combined Modality Therapy , Follow-Up Studies , Genetic Vectors , Humans , Infant , Lymphocyte Count , Retroviridae , Severe Combined Immunodeficiency/immunology , Transduction, Genetic , Transplantation Conditioning
11.
J Immunol ; 180(7): 4415-24, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18354162

ABSTRACT

Conventional MHC-restricted T lymphocytes leave thymus with a naive phenotype and require Ag-dependent stimulation coupled to proliferation to acquire effector functions. Invariant (i)NKT cells are a subset of T lymphocytes considered innate because they display an effector memory phenotype independent of TCR stimulation by foreign Ags. We investigated the effector differentiation program followed by human iNKT cells by studying cells from a relevant set of fetal thymi and umbilical cord blood samples. We find that human fetal iNKT cells have already started a differentiation program that activates the epigenetic and transcriptional control of ifng and il4 genes, leading at birth to cells that express these cytokines upon TCR signaling but independently of proliferation in vitro. Both ex vivo and in vitro analysis of fetal and neonatal iNKT cells delineate an effector differentiation program linked to cell division in vivo, and they identify IL-7 as one of the crucial signals driving this program in the apparent absence of Ag stimulation. Consistent with these data, human fetal and neonatal iNKT cells are hyperresponsive in vitro to IL-7 in comparison to conventional T cells, owing to an increased expression and signaling function of the IL-7 receptor alpha-chain. The innate nature of human iNKT cells could thus derive from lineage-specific developmental cues that selectively make these cells efficient IL-7 responders following thymic selection.


Subject(s)
Cell Differentiation/drug effects , Cell Differentiation/immunology , Immunity, Innate/drug effects , Immunity, Innate/immunology , Interleukin-7/pharmacology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Epigenesis, Genetic/genetics , Epigenesis, Genetic/immunology , Gene Expression Regulation , Humans , Infant, Newborn , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Time Factors , Transcription, Genetic/genetics
12.
J Clin Invest ; 117(8): 2233-40, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17671653

ABSTRACT

Gene transfer into HSCs is an effective treatment for SCID, although potentially limited by the risk of insertional mutagenesis. We performed a genome-wide analysis of retroviral vector integrations in genetically corrected HSCs and their multilineage progeny before and up to 47 months after transplantation into 5 patients with adenosine deaminase-deficient SCID. Gene-dense regions, promoters, and transcriptionally active genes were preferred retroviral integrations sites (RISs) both in preinfusion transduced CD34(+) cells and in vivo after gene therapy. The occurrence of insertion sites proximal to protooncogenes or genes controlling cell growth and self renewal, including LMO2, was not associated with clonal selection or expansion in vivo. Clonal analysis of long-term repopulating cell progeny in vivo revealed highly polyclonal T cell populations and shared RISs among multiple lineages, demonstrating the engraftment of multipotent HSCs. These data have important implications for the biology of retroviral vectors, the dynamics of genetically modified HSCs, and the safety of gene therapy.


Subject(s)
Adenosine Deaminase , Genetic Therapy , Hematopoietic Stem Cell Transplantation , Mutagenesis, Insertional , Retroviridae , Severe Combined Immunodeficiency/therapy , Virus Integration/genetics , Adaptor Proteins, Signal Transducing , Adenosine Deaminase/genetics , Antigens, CD34 , Child, Preschool , DNA-Binding Proteins/genetics , Female , Hematopoietic Stem Cells/metabolism , Humans , Infant , LIM Domain Proteins , Male , Metalloproteins/genetics , Multipotent Stem Cells/metabolism , Proto-Oncogene Proteins , Risk Factors , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/metabolism , T-Lymphocytes/metabolism , Transplantation, Autologous
13.
J Immunol ; 174(8): 5074-81, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15817443

ABSTRACT

In patients with primary Ab deficiencies, hematological and immunological abnormalities are frequently observed. A regenerative failure of hemopoietic stem/progenitor cells has been hypothesized. We evaluated in the bone marrow (BM) of 11 patients with common variable immunodeficiency, the phenotype of BM progenitors and their in vitro growth by colony-forming cell (CFC) and long-term culture (LTC) assays. A significant decrease in erythroid and mixed CFC and, to a greater extent, in primitive LTC-CFC progenitors was observed in patients compared with healthy controls. The frequency of BM pre-B and pro-B cells correlated directly with the absolute number of CD19+ lymphocytes. BM cells cultured in vitro produced spontaneously lower amounts of IL-2 and elevated levels of TNF-alpha compared with controls, indicating a skewing toward a proapoptotic cytokine pattern. In addition, stromal cells generated after BM LTC secreted less IL-7 and displayed by immunohistochemistry an altered phenotype. These findings were associated with a significant decrease in naive Th cells coexpressing CD31 in the peripheral blood. These results indicate an impaired growth and differentiation capacity of progenitor cells in patients with common variable immunodeficiency.


Subject(s)
Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Common Variable Immunodeficiency/immunology , Common Variable Immunodeficiency/pathology , Cytokines/biosynthesis , Thymus Gland/immunology , Thymus Gland/pathology , Adult , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Case-Control Studies , Colony-Forming Units Assay , Common Variable Immunodeficiency/blood , Female , Hematopoiesis , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Humans , In Vitro Techniques , Interleukin-2/biosynthesis , Interleukin-7/biosynthesis , Male , Middle Aged , Tumor Necrosis Factor-alpha/biosynthesis
14.
J Clin Invest ; 115(4): 1049-59, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15761493

ABSTRACT

X-linked lymphoproliferative disease (XLP) is an often-fatal immunodeficiency characterized by hypogammaglobulinemia, fulminant infectious mononucleosis, and/or lymphoma. The genetic lesion in XLP, SH2D1A, encodes the adaptor protein SAP (signaling lymphocytic activation molecule-associated [SLAM-associated] protein); however, the mechanism(s) by which mutations in SH2D1A causes hypogammaglobulinemia is unknown. Our analysis of 14 XLP patients revealed normal B cell development but a marked reduction in the number of memory B cells. The few memory cells detected were IgM(+), revealing deficient isotype switching in vivo. However, XLP B cells underwent proliferation and differentiation in vitro as efficiently as control B cells, which indicates that the block in differentiation in vivo is B cell extrinsic. This possibility is supported by the finding that XLP CD4(+) T cells did not efficiently differentiate into IL-10(+) effector cells or provide optimal B cell help in vitro. Importantly, the B cell help provided by SAP-deficient CD4(+) T cells was improved by provision of exogenous IL-10 or ectopic expression of SAP, which resulted in increased IL-10 production by T cells. XLP CD4(+) T cells also failed to efficiently upregulate expression of inducible costimulator (ICOS), a potent inducer of IL-10 production by CD4(+) T cells. Thus, insufficient IL-10 production may contribute to hypogammaglobulinemia in XLP. This finding suggests new strategies for treating this immunodeficiency.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Interleukin-10/biosynthesis , Lymphoproliferative Disorders/immunology , Adolescent , Adult , Agammaglobulinemia/genetics , Agammaglobulinemia/metabolism , Animals , Antibody Formation , Antigens, Differentiation, T-Lymphocyte/immunology , B-Lymphocytes/immunology , CD40 Antigens/immunology , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Child , Humans , Immunoglobulin Class Switching , Immunologic Memory , Inducible T-Cell Co-Stimulator Protein , Intracellular Signaling Peptides and Proteins/metabolism , Lymphoproliferative Disorders/genetics , Middle Aged , Signaling Lymphocytic Activation Molecule Associated Protein
15.
Blood ; 105(11): 4383-9, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15677558

ABSTRACT

The adaptor protein SAP regulates signaling through signaling lymphocytic activation molecule (SLAM)-family receptors expressed on T and natural killer (NK) cells. In patients affected by X-linked lymphoproliferative (XLP) disease, mutations in the SH2D1A gene result in defective lytic activity. However, the mechanism by which SAP controls cytotoxic activity remains unclear. T-cell-receptor (TCR) activation of CD8(+) cytotoxic T cells (CTLs) results in down-regulation of SAP, suggesting that this protein is involved in early activation events. Here, we show that SAP-deficient CTLs from patients with XLP and hemophagocytic lymphohistiocytosis (HLH) display a specific lytic defect against autologous and allogeneic Epstein-Barr virus (EBV)-positive B cells. This defect is associated with the defective polarization of 2B4, perforin, and lipid rafts at the contact area of CTLs with EBV-positive targets. Blockade of 2B4 in normal CTLs reproduces the defects in lysis and polarization observed in SAP-deficient CTLs. Expression and regulation of the SLAM-family receptors SLAM, CD84, and 2B4, as well as the lytic effectors perforin and granzyme-B are normal in SAP-deficient CTLs. In addition, TCR stimulation leads to normal proliferation and production of interleukin 2 (IL-2), IL-4, and interferon-gamma (IFN-gamma). These results demonstrate that the SAP/2B4 pathway plays a key role in CTL lytic activity against EBV-positive targets by promoting the polarization of the lytic machinery.


Subject(s)
B-Lymphocytes/virology , CD8-Positive T-Lymphocytes/immunology , Cytotoxicity, Immunologic , Epstein-Barr Virus Infections/immunology , Intracellular Signaling Peptides and Proteins/physiology , Antigens, CD/metabolism , B-Lymphocytes/immunology , Cell Communication , Epstein-Barr Virus Infections/pathology , Granzymes , Histiocytosis, Non-Langerhans-Cell/genetics , Histiocytosis, Non-Langerhans-Cell/immunology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/immunology , Membrane Glycoproteins/metabolism , Membrane Microdomains/metabolism , Receptors, Immunologic/metabolism , Serine Endopeptidases/metabolism , Signaling Lymphocytic Activation Molecule Associated Protein , Signaling Lymphocytic Activation Molecule Family
16.
Mol Ther ; 10(6): 1096-108, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15564141

ABSTRACT

To improve maintenance and gene transfer of human lymphoid progenitors for clinical use in gene therapy of adenosine deaminase (ADA)-deficient SCID we investigated several gene transfer protocols using various stem cell-enriched sources. The lymphoid differentiation potential was measured by an in vitro clonal assay for B/NK cells and in the in vivo SCID-hu mouse model. Ex vivo culture with the cytokines TPO, FLT3-ligand, and SCF (T/F/S) plus IL-3 or IL-7 substantially increased the yield of transduced bone marrow (BM) CD34(+) cells purified from ADA-SCID patients or healthy donors, compared to T/F/S alone. Moreover, the use of IL-3 or IL-7 significantly improved the maintenance of in vitro B cell progenitors from ADA-SCID BM cells and allowed the efficient transduction of B and NK cell progenitors. Under these optimized conditions transduced CD34(+) cells were efficiently engrafted into SCID-hu mice and gave rise to B and T cell progeny, demonstrating the maintenance of in vivo lymphoid reconstitution capacity. The protocol based on the T/F/S + IL-3 combination was included in a gene therapy clinical trial for ADA-SCID, resulting in long-term engraftment of stem/progenitor cells. Remarkably, gene-corrected BM CD34(+) cells obtained from one patient 4 and 11 months after gene therapy were capable of repopulating the lymphoid compartment of SCID-hu hosts.


Subject(s)
Adenosine Deaminase/metabolism , Antigens, CD34/metabolism , Bone Marrow Cells/drug effects , Gene Transfer Techniques , Interleukin-3/pharmacology , Interleukin-7/pharmacology , Lymphocytes/drug effects , Severe Combined Immunodeficiency/pathology , Adenosine Deaminase/deficiency , Adenosine Deaminase/genetics , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Differentiation , Cell Proliferation/drug effects , Cells, Cultured , Fetal Blood/drug effects , Fetal Blood/metabolism , Genetic Therapy , Humans , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Mice, SCID , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/metabolism , Severe Combined Immunodeficiency/therapy , Stem Cell Transplantation , Transduction, Genetic
17.
Am J Med Genet A ; 120A(2): 222-8, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12833403

ABSTRACT

Opitz (or G/BBB) syndrome is a pleiotropic genetic disorder characterized by hypertelorism, hypospadias, and additional midline defects. This syndrome is heterogeneous with an X-linked (XLOS) and an autosomal dominant (ADOS) form. The gene implicated in the XLOS form, MID1, encodes a protein containing a RING-Bbox-Coiled-coil motif belonging to the tripartite motif (TRIM) family. To further clarify the molecular basis of XLOS, we have undertaken mutation analysis of the MID1 gene in patients with Opitz syndrome (OS). We found novel mutations in 11 of 63 male individuals referred to us as sporadic or familial X-linked OS cases. The mutations are scattered throughout the gene, although more are represented in the 3' region. By reviewing all the MID1-mutated OS patients so far described, we confirmed that hypertelorism and hypospadias are the most frequent manifestations, being present in almost every XLOS individual. However, it is clear that laryngo-tracheo-esophageal (LTE) defects are also common anomalies, being manifested by all MID1-mutated male patients. Congenital heart and anal abnormalities are less frequent than reported in literature. In addition, we can include limb defects in the OS clinical synopsis as we found a MID1-mutated patient showing syndactyly. The low frequency of mutations in MID1 and the high variability of the phenotype suggest the involvement of other genes in the OS phenotype.


Subject(s)
Abnormalities, Multiple/genetics , Chromosomes, Human, X/genetics , Mutation , 3' Flanking Region , Abnormalities, Multiple/pathology , DNA Mutational Analysis , Genetic Linkage , Humans , Hypertelorism/genetics , Hypertelorism/pathology , Hypospadias/genetics , Hypospadias/pathology , Larynx/abnormalities , Male , Pedigree , Syndrome
18.
Science ; 296(5577): 2410-3, 2002 Jun 28.
Article in English | MEDLINE | ID: mdl-12089448

ABSTRACT

Hematopoietic stem cell (HSC) gene therapy for adenosine deaminase (ADA)-deficient severe combined immunodeficiency (SCID) has shown limited clinical efficacy because of the small proportion of engrafted genetically corrected HSCs. We describe an improved protocol for gene transfer into HSCs associated with nonmyeloablative conditioning. This protocol was used in two patients for whom enzyme replacement therapy was not available, which allowed the effect of gene therapy alone to be evaluated. Sustained engraftment of engineered HSCs with differentiation into multiple lineages resulted in increased lymphocyte counts, improved immune functions (including antigen-specific responses), and lower toxic metabolites. Both patients are currently at home and clinically well, with normal growth and development. These results indicate the safety and efficacy of HSC gene therapy combined with nonmyeloablative conditioning for the treatment of SCID.


Subject(s)
Adenosine Deaminase/deficiency , Adenosine Deaminase/genetics , Genetic Therapy , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Severe Combined Immunodeficiency/therapy , Transplantation Conditioning , Adenosine Deaminase/metabolism , Animals , B-Lymphocytes/enzymology , B-Lymphocytes/immunology , Bone Marrow Transplantation , Cell Differentiation , Child, Preschool , Genetic Vectors , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Humans , Immunoglobulins/blood , Infant , Leukocytes/enzymology , Leukopoiesis , Lymphocyte Activation , Mice , Mice, SCID , Retroviridae/genetics , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...