Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Gene Ther ; 23(11): 815-818, 2016 11.
Article in English | MEDLINE | ID: mdl-27487944

ABSTRACT

Integrating vectors based on γ-retroviruses and containing full-length long terminal repeats (LTRs) have been associated with activation of oncogene expression and leukemogenesis in human gene therapy trials. Identification of the specific molecular elements of the LTRs that have a role in insertional oncogenesis events is important as it can lead to the development of safer gene transfer vectors. The negative control region (NCR) of the LTR is a particularly well-conserved sequence among mammalian γ-retroviruses with demonstrated regulatory activity of gene transcription in hematopoietic cells, which led us to hypothesize that this region may have a role in insertional oncogenesis after γ-retroviral vector (GV)-mediated gene transfer into hematopoietic progenitors. We used an in vitro assay of murine bone marrow cell immortalization to compare the immortalization capabilities of a series of GVs carrying murine leukemia virus (MLV) LTR deletion mutants. Compared with GV carrying the full-length MLV LTR, deletion of the complete LTR enhancer sequence showed significant reduction of immortalization rates. However, the use of a mutant LTR deleted of the enhancer sequence, with exception of the NCR, did not affect immortalization. Importantly, the inclusion of an LTR mutant devoid only of the NCR did show significant reduction of immortalization rates compared with the full LTR sequence. Therefore, our data point to the NCR as a key element for immortalization and justify additional studies to evaluate its specific role in MLV-mediated insertional oncogenesis.


Subject(s)
Cell Transformation, Viral , Genetic Vectors/genetics , Hematopoietic Stem Cells/metabolism , Moloney murine leukemia virus/genetics , Terminal Repeat Sequences , Animals , Cells, Cultured , Gene Transfer Techniques , Mice , Mice, Inbred C57BL , Mutagenesis, Insertional
2.
N Engl J Med ; 371(6): 507-518, 2014 08 07.
Article in English | MEDLINE | ID: mdl-25029335

ABSTRACT

BACKGROUND: The study of autoinflammatory diseases has uncovered mechanisms underlying cytokine dysregulation and inflammation. METHODS: We analyzed the DNA of an index patient with early-onset systemic inflammation, cutaneous vasculopathy, and pulmonary inflammation. We sequenced a candidate gene, TMEM173, encoding the stimulator of interferon genes (STING), in this patient and in five unrelated children with similar clinical phenotypes. Four children were evaluated clinically and immunologically. With the STING ligand cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), we stimulated peripheral-blood mononuclear cells and fibroblasts from patients and controls, as well as commercially obtained endothelial cells, and then assayed transcription of IFNB1, the gene encoding interferon-ß, in the stimulated cells. We analyzed IFNB1 reporter levels in HEK293T cells cotransfected with mutant or nonmutant STING constructs. Mutant STING leads to increased phosphorylation of signal transducer and activator of transcription 1 (STAT1), so we tested the effect of Janus kinase (JAK) inhibitors on STAT1 phosphorylation in lymphocytes from the affected children and controls. RESULTS: We identified three mutations in exon 5 of TMEM173 in the six patients. Elevated transcription of IFNB1 and other gene targets of STING in peripheral-blood mononuclear cells from the patients indicated constitutive activation of the pathway that cannot be further up-regulated with stimulation. On stimulation with cGAMP, fibroblasts from the patients showed increased transcription of IFNB1 but not of the genes encoding interleukin-1 (IL1), interleukin-6 (IL6), or tumor necrosis factor (TNF). HEK293T cells transfected with mutant constructs show elevated IFNB1 reporter levels. STING is expressed in endothelial cells, and exposure of these cells to cGAMP resulted in endothelial activation and apoptosis. Constitutive up-regulation of phosphorylated STAT1 in patients' lymphocytes was reduced by JAK inhibitors. CONCLUSIONS: STING-associated vasculopathy with onset in infancy (SAVI) is an autoinflammatory disease caused by gain-of-function mutations in TMEM173. (Funded by the Intramural Research Program of the National Institute of Arthritis and Musculoskeletal and Skin Diseases; ClinicalTrials.gov number, NCT00059748.).


Subject(s)
Inflammation/genetics , Membrane Proteins/genetics , Mutation , Skin Diseases, Vascular/genetics , Age of Onset , Cytokines/genetics , Cytokines/metabolism , Female , Fibroblasts/metabolism , Genes, Dominant , Humans , Infant , Infant, Newborn , Inflammation/metabolism , Interferon-gamma/genetics , Interferon-gamma/metabolism , Janus Kinases/antagonists & inhibitors , Lung Diseases/genetics , Male , Pedigree , Phosphorylation , STAT1 Transcription Factor/metabolism , Sequence Analysis, DNA , Skin Diseases, Vascular/metabolism , Syndrome , Transcription, Genetic , Up-Regulation
3.
Clin Immunol ; 142(2): 160-6, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22079330

ABSTRACT

Wiskott-Aldrich syndrome (WAS) is a rare X-linked disorder caused by mutations in the WAS gene. Glomerulonephritis is a frequent complication, however, histopathological data from affected patients is scarce because the thrombocytopenia that affects most patients is a contraindication to renal biopsies. We found that WASp-deficient mice develop proliferative glomerulonephritis reminiscent of human IgA nephropathy (IgAN). We examined whether increased aberrant IgA production is associated with the development of glomerulonephritis in WASp-deficient mice. Serum IgA and IgA production by splenic B cells was increased in WASp-deficient mice compared to wild-type (WT) mice. A lectin-binding study revealed a reduced ratio of sialylated and galactosylated IgA in the sera from old WASp-deficient mice. Circulating IgA-containing immune complexes showed significantly higher titers in WASp-deficient mice compared to WT mice. These results indicate that the increased IgA production and aberrant glycosylation of IgA may be critically involved in the pathogenesis of glomerulonephritis in WAS.


Subject(s)
Glomerulonephritis, IGA/immunology , Immunoglobulin A/metabolism , Wiskott-Aldrich Syndrome Protein/deficiency , Wiskott-Aldrich Syndrome/immunology , Animals , B-Lymphocytes/immunology , Disease Models, Animal , Glomerulonephritis, IGA/metabolism , Glomerulonephritis, IGA/pathology , Glycosylation , Humans , Immunoglobulin A/blood , Mice , Mice, Knockout , Spleen/immunology , Thrombocytopenia/metabolism
4.
Scand J Immunol ; 74(5): 471-81, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21671975

ABSTRACT

Patients with adenosine deaminase (ADA) deficiency exhibit spontaneous and partial clinical remission associated with somatic reversion of inherited mutations. We report a child with severe combined immunodeficiency (T-B- SCID) due to ADA deficiency diagnosed at the age of 1 month, whose lymphocyte counts including CD4+ and CD8+ T and NK cells began to improve after several months with normalization of ADA activity in Peripheral blood lymphocytes (PBL), as a result of somatic mosaicism caused by monoallelic reversion of the causative mutation in the ADA gene. He was not eligible for haematopoietic stem cell transplantation (HSCT) or gene therapy (GT); therefore he was placed on enzyme replacement therapy (ERT) with bovine PEG-ADA. The follow-up of metabolic and immunologic responses to ERT included gradual improvement in ADA activity in erythrocytes and transient expansion of most lymphocyte subsets, followed by gradual stabilization of CD4+ and CD8+ T (with naïve phenotype) and NK cells, and sustained expansion of TCRγδ+ T cells. This was accompanied by the disappearance of the revertant T cells as shown by DNA sequencing from PBL. Although the patient's clinical condition improved marginally, he later developed a germinal cell tumour and eventually died at the age of 67 months from sepsis. This case adds to our current knowledge of spontaneous reversion of mutations in ADA deficiency and shows that the effects of the ERT may vary among these patients, suggesting that it could depend on the cell and type in which the somatic mosaicism is established upon reversion.


Subject(s)
Adenosine Deaminase/metabolism , Enzyme Replacement Therapy , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Neoplasms, Unknown Primary/genetics , Neoplasms, Unknown Primary/therapy , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/therapy , Adenosine Deaminase/administration & dosage , Adenosine Deaminase/genetics , Adenosine Deaminase/immunology , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cattle , Cell Count , Child , Child, Preschool , DNA Mutational Analysis , Fatal Outcome , Humans , Immunophenotyping , Infant , Killer Cells, Natural/pathology , Lung Neoplasms/complications , Lung Neoplasms/physiopathology , Lung Neoplasms/secondary , Male , Mosaicism/drug effects , Mutation/genetics , Neoplasms, Unknown Primary/complications , Neoplasms, Unknown Primary/pathology , Neoplasms, Unknown Primary/physiopathology , Receptors, Antigen, T-Cell/metabolism , Severe Combined Immunodeficiency/complications , Severe Combined Immunodeficiency/pathology , Severe Combined Immunodeficiency/physiopathology , Shock, Septic
6.
Mol Cell ; 8(5): 959-69, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11741532

ABSTRACT

Janus kinases comprise carboxyterminal kinase, pseudokinase, SH2-like, and N-terminal FERM domains. We identified three patient-derived mutations in the FERM domain of Jak3 and investigated the functional consequences of these mutations. These mutations inhibited receptor binding and also abrogated kinase activity, suggesting interactions between the FERM and kinase domains. In fact, the domains were found to physically associate, and coexpression of the FERM domain enhanced activity of the isolated kinase domain. Conversely, staurosporine, which alters kinase domain structure, disrupted receptor binding, even though the catalytic activity of Jak3 is dispensable for receptor binding. Thus, the Jak FERM domain appears to have two critical functions: receptor interaction and maintenance of kinase integrity.


Subject(s)
Protein-Tyrosine Kinases/metabolism , Adenosine Triphosphate/analogs & derivatives , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , COS Cells , Catalysis , Enzyme Inhibitors/pharmacology , Humans , Interleukin Receptor Common gamma Subunit , Janus Kinase 3 , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Protein Structure, Tertiary , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/genetics , Pyrimidines/pharmacology , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Staurosporine/pharmacology , src-Family Kinases/antagonists & inhibitors
7.
Biochem Biophys Res Commun ; 289(2): 525-30, 2001 Nov 30.
Article in English | MEDLINE | ID: mdl-11716505

ABSTRACT

A method is described for the preparation of ganciclovir triphosphate (GCV-TP) using murine colon cancer cells (MC38) transduced with the herpes simplex virus-thymidine kinase (MC38/HSV-tk). Murine cells transduced with viral-tk contain required viral and host enzymes needed for complete cellular synthesis of this potent antiviral metabolite. Dose response studies showed optimal intracellular levels of GCV-TP occurred after exposure of MC38/HSV-tk cells to 300 microM ganciclovir for 24 h producing 7.5 nmol GCV-TP/10(6) cells. This reflects cellular accumulation of GCV-TP to levels 25-fold greater than the medium concentration of parent drug. A simple isolation scheme included methanolic extraction and anion-exchange chromatography to recover the target triphosphate. Mass spectral analysis and selective enzyme degradation provided structural confirmation of the purified product. Biological activity of the purified GCV-TP was demonstrated by competitive inhibition experiments using human DNA polymerase alpha and HSV DNA polymerase that showed substantially greater sensitivity for the viral polymerase in agreement with previous reports. The GCV-TP obtained was further used to enzymatically prepare GCV mono- and diphosphate in high yield. This method provides an easily scalable means of preparing milligram amounts of the triphosphates of pharmacologically active acyclic nucleosides like ganciclovir.


Subject(s)
Ganciclovir/analogs & derivatives , Ganciclovir/chemistry , Ganciclovir/isolation & purification , Ganciclovir/pharmacology , Adenocarcinoma/metabolism , Animals , Anions , Antiviral Agents/pharmacology , Binding, Competitive , Cells, Cultured , Chromatography, High Pressure Liquid , Chromatography, Ion Exchange , Colonic Neoplasms/metabolism , DNA Polymerase I/metabolism , DNA-Directed DNA Polymerase/metabolism , Dose-Response Relationship, Drug , Herpes Simplex/enzymology , Humans , Kinetics , Mass Spectrometry , Mice , Models, Chemical , Thymidine Kinase/metabolism , Time Factors , Transduction, Genetic , Tumor Cells, Cultured
8.
Proc Natl Acad Sci U S A ; 98(15): 8697-702, 2001 Jul 17.
Article in English | MEDLINE | ID: mdl-11447283

ABSTRACT

Somatic mosaicism caused by in vivo reversion of inherited mutations has been described in several human genetic disorders. Back mutations resulting in restoration of wild-type sequences and second-site mutations leading to compensatory changes have been shown in mosaic individuals. In most cases, however, the precise genetic mechanisms underlying the reversion events have remained unclear, except for the few instances where crossing over or gene conversion have been demonstrated. Here, we report a patient affected with Wiskott--Aldrich syndrome (WAS) caused by a 6-bp insertion (ACGAGG) in the WAS protein gene, which abrogates protein expression. Somatic mosaicism was documented in this patient whose majority of T lymphocytes expressed nearly normal levels of WAS protein. These lymphocytes were found to lack the deleterious mutation and showed a selective growth advantage in vivo. Analysis of the sequence surrounding the mutation site showed that the 6-bp insertion followed a tandem repeat of the same six nucleotides. These findings strongly suggest that DNA polymerase slippage was the cause of the original germ-line insertion mutation in this family and that the same mechanism was responsible for its deletion in one of the propositus T cell progenitors, thus leading to reversion mosaicism.


Subject(s)
Mosaicism , Proteins/genetics , Wiskott-Aldrich Syndrome/genetics , Adult , CD3 Complex/biosynthesis , Complementarity Determining Regions/genetics , Female , Humans , Male , Mutagenesis, Insertional , Pedigree , Protein Biosynthesis , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Wiskott-Aldrich Syndrome/immunology , Wiskott-Aldrich Syndrome Protein
9.
Cell Transplant ; 10(1): 81-9, 2001.
Article in English | MEDLINE | ID: mdl-11294475

ABSTRACT

Ex vivo embryonic liver explant culture is a novel and attractive approach to obtain abundant hepatic and hematopoietic stem cells. Gene therapy of autologous hepatic and hematopoietic stem cells represents an alternative therapeutic approach to liver transplantation for genetic and metabolic disorders. In this study we characterize the growth and differentiation of hepatic stem cells utilizing embryonic liver cultures. Day 9.5 liver buds are microdissected and cultured under specific conditions. Modulation of growth conditions by addition of hepatocyte growth factor, Flt-3 ligand, and stem cell factor leads to enrichment of hepatic progenitor cells in embryonic liver explants. Under these conditions, we also demonstrate the role of a novel marker PRAJA-1 to identify hepatic stem cells and transitional hepatocytes. Utilization of dexamethasone enhanced pseudolobule formation with increased hepatocytic and biliary differentiation. Transforming growth factor-beta leads to enrichment of biliary cells in the culture. Gut formation is enhanced in the presence of interleukin-3 and blood formation by increasing the mesodermal tissue in these cultures. We also show increased retroviral-mediated expression of the green fluorescent protein expression in the expanded hepatic and hematopoietic stem cells under different culture conditions. Thus, the embryonic liver explant culture is an attractive source for hepatic progenitors and is a possible step towards generating nontumorigenic immortalized hepatocytes with possible transplantation applications.


Subject(s)
Fetal Tissue Transplantation/methods , Hematopoietic Stem Cells/cytology , Hepatocytes/cytology , Liver Transplantation/methods , Animals , Biliary Tract/cytology , Cell Differentiation/drug effects , Female , Fetal Tissue Transplantation/pathology , Genetic Therapy , Hepatocyte Growth Factor/pharmacology , Hepatocytes/drug effects , Hepatocytes/transplantation , Liver Transplantation/pathology , Membrane Proteins/pharmacology , Mice , Mice, Inbred ICR , Phenotype , Pregnancy , Stem Cell Factor/pharmacology , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/drug effects
10.
Mol Ther ; 3(4): 565-73, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11319919

ABSTRACT

X-linked severe combined immunodeficiency (XSCID) is caused by mutations in the IL-2 receptor gamma chain (IL2RG) gene, resulting in absent T lymphocytes and nonfunctional B lymphocytes. Recently T lymphocyte production and B lymphocyte function were restored in XSCID patients infused with autologous stem cells transduced with a retrovirus containing the human IL2RG cDNA. To optimize the expression of human IL2RG for future clinical trials, we compared five retroviral vectors expressing human IL2RG from different LTR enhancer-promoter elements in a mouse model. Northern and Southern blot analysis of hematopoietic tissues from repopulated mice revealed that the retroviral vector with the highest expression per copy number was MFG-S-hIL2RG, followed by MND-hIL2RG. All five vectors were capable of restoring lymphopoiesis in irradiated XSCID mice transplanted with transduced IL2RG-deficient hematopoietic stem cells. Transduction of IL2RG-deficient hematopoietic stem cells with all five vectors restored T lymphopoiesis in transplanted stem cell-deficient W/W(v) mouse recipients. However, only XSCID stem cells transduced with the MFG-S-hIL2RG vector generated B lymphocytes in W/W(v) mice. We conclude that the MFG-S-hIL2RG vector provides the best opportunity for in vivo selection and development of B and T lymphocytes for human XSCID gene therapy.


Subject(s)
B-Lymphocytes/metabolism , Genetic Linkage , Genetic Vectors , Receptors, Interleukin-2/genetics , Retroviridae/genetics , Severe Combined Immunodeficiency/therapy , T-Lymphocytes/metabolism , X Chromosome/genetics , 3T3 Cells , Animals , Blotting, Northern , Blotting, Southern , DNA, Complementary/metabolism , Disease Models, Animal , Female , Flow Cytometry , Genetic Therapy/methods , Hematopoietic Stem Cells/metabolism , Humans , Lymphocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Models, Genetic , Mutation , Promoter Regions, Genetic , RNA, Messenger/metabolism , Retroviridae/metabolism , Time Factors , Transduction, Genetic
11.
Blood ; 97(6): 1618-24, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11238100

ABSTRACT

A recent clinical trial of gene therapy for X-linked severe combined immunodeficiency (XSCID) has shown that retroviral-mediated gene correction of bone marrow stem cells can lead to the development of normal immune function. These exciting results have been preceded by successful immune reconstitution in several XSCID mouse models, all carrying null mutations of the common gamma chain (gamma(c)). One question not formally addressed by these previous studies is that of possible dominant-negative effects of the endogenous mutant gamma(c) protein on the activity of the wild-type transferred gene product. The present work was therefore undertaken to study whether corrective gene transfer was applicable to an XSCID murine model with preserved expression of a truncated gammac molecule (Deltagamma(c+)-XSCID). Gene correction of Deltagamma(c+)-XSCID mice resulted in the reconstitution of lymphoid development, and preferential repopulation of lymphoid organs by gene-corrected cells demonstrated the selective advantage of gamma(c)-expressing cells in vivo. Newly developed B cells showed normalization of lipopolysaccharide-mediated proliferation and interleukin-4 (IL-4)-induced immunoglobulin G1 isotype switching. Splenic T cells and thymocytes of treated animals proliferated normally to mitogens and responded to the addition of IL-2, IL-4, and IL-7, indicating functional reconstitution of gammac-sharing receptors. Repopulated thymi showed a clear increase of CD4-/CD8- and CD8+ fractions, both dramatically reduced in untreated Deltagamma(c+)-XSCID mice. These improvements were associated with the restoration of Bcl-2 expression levels and enhanced cell survival. These data indicate that residual expression of the endogenous truncated gamma(c) did not lead to dominant-negative effects in this murine model and suggest that patient selection may not be strictly necessary for gene therapy of XSCID.


Subject(s)
Genetic Therapy/methods , Immunoglobulin gamma-Chains/therapeutic use , Mice, SCID/immunology , Animals , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Cell Survival/drug effects , Genes, Dominant , Genetic Therapy/standards , Hematopoietic Stem Cells/metabolism , Immunoglobulin Class Switching/drug effects , Immunoglobulin gamma-Chains/genetics , Immunoglobulin gamma-Chains/metabolism , Lymphocyte Activation/drug effects , Lymphocytes/cytology , Lymphocytes/drug effects , Mice , Proto-Oncogene Proteins c-bcl-2/metabolism , Retroviridae/genetics , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/metabolism , Transduction, Genetic
12.
Curr Allergy Asthma Rep ; 1(5): 407-15, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11892066

ABSTRACT

Since the early 1990s, primary immunodeficiency (ID) disorders have played a major role in the development of human gene therapy. Adenosine deaminase (ADA) deficiency was the first disease to be treated with a gene therapy approach in humans, and was also the first condition for which therapeutic gene transfer into the hematopoietic stem cell has been attempted in the clinical arena. A series of encouraging results obtained in chronic granulomatous disease (CGD) patients have followed these pioneer experiments and preceded the very recent and exciting reports of successful genetic correction procedures performed in patients affected with the X-linked form of severe combined immunodeficiency (XSCID). The technical progress made in the field of gene transfer in recent years is mostly responsible for these clinical advances, and will be critical for future development of gene therapy approaches for other forms of IDs.


Subject(s)
Genetic Therapy , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/therapy , Adenosine Deaminase/deficiency , Adenosine Deaminase/genetics , Humans
13.
Curr Opin Allergy Clin Immunol ; 1(6): 497-501, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11964732

ABSTRACT

Primary immunodeficiency diseases have been important targets of corrective gene transfer approaches since the very early days of gene therapy. The potential for selective survival advantage of gene-corrected cells over populations carrying the mutated, causative gene translates into the possibility of obtaining clinical meaningful results in patients with primary immunodeficiency diseases even if levels of gene transfer are low. This critical prospect has fueled the interest of researchers since the mid-1980s and has recently determined the success of a clinical trial of gene therapy for X-linked severe combined immunodeficiency.


Subject(s)
Genetic Therapy/methods , Immunologic Deficiency Syndromes/therapy , Animals , Humans , Leukocyte-Adhesion Deficiency Syndrome/therapy , Mice , Severe Combined Immunodeficiency/therapy , X Chromosome
14.
Genes Immun ; 2(8): 422-32, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11781709

ABSTRACT

Mutations of the Janus kinase 3 (JAK3) have been previously described to cause an autosomal recessive variant of severe combined immunodeficiency (SCID) usually characterized by the near absence of T and NK cells, but preserved numbers of B lymphocytes (T-B+SCID). We now report a family whose JAK3 mutations are associated with the persistence of circulating T cells, resulting in previously undescribed clinical presentations, ranging from a nearly unaffected 18-year-old subject to an 8-year-old sibling with a severe lymphoproliferative disorder. Both siblings were found to be compound heterozygotes for the same deleterious JAK3 mutations: an A96G initiation start site mutation, resulting in a dysfunctional, truncated protein product and a G2775(+3)C mutation in the splice donor site sequence of intron 18, resulting in a splicing defect and a predicted premature stop. These mutations were compatible with minimal amounts of functional JAK3 expression, leading to defective cytokine-dependent signaling. Activated T cells in these patients failed to express Fas ligand (FasL) in response to IL-2, which may explain the accumulation of T cells with an activated phenotype and a skewed T cell receptor (TcR) Vbeta family distribution. We speculate that residual JAK3 activity accounted for the maturation of thymocytes, but was insufficient to sustain IL-2-mediated homeostasis of peripheral T cells via Fas/FasL interactions. These data demonstrate that the clinical spectrum of JAK3 deficiency is quite broad and includes immunodeficient patients with accumulation of activated T cells, and indicate an essential role for JAK3 in the homeostasis of peripheral T cells in humans.


Subject(s)
Protein-Tyrosine Kinases/deficiency , Adolescent , Amino Acid Sequence , B-Lymphocytes/metabolism , Cell Line, Transformed , Child , DNA, Complementary , Fas Ligand Protein , Female , Gene Expression , Humans , Immunophenotyping , Interleukin-2/metabolism , Janus Kinase 3 , Male , Membrane Glycoproteins/metabolism , Molecular Sequence Data , Mutation , Pedigree , Protein-Tyrosine Kinases/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Interleukin-2/metabolism , Sequence Analysis, DNA , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/immunology , Severe Combined Immunodeficiency/pathology , Signal Transduction , T-Lymphocytes , Up-Regulation
15.
Pediatr Clin North Am ; 47(6): 1389-407, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11131002

ABSTRACT

Several hurdles remain before gene therapy will be a part of mainstream medical therapy; however, the preliminary report of success in HSC correction in patients with XSCID provides hope that gene therapy will become a reality.


Subject(s)
Genetic Therapy/methods , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/therapy , Animals , Child, Preschool , Female , Humans , Immunologic Deficiency Syndromes/diagnosis , Infant , Infant, Newborn , Male , Prognosis , Treatment Outcome
16.
Hum Gene Ther ; 11(14): 2051-6, 2000 Sep 20.
Article in English | MEDLINE | ID: mdl-11020803

ABSTRACT

Corrective gene transfer into hematopoietic stem cells (HSCs) is being investigated as therapy for X-linked severe combined immunodeficiency (XSCID) and it is hoped that selective advantage of gene-corrected HSCs will help in achieving full immune reconstitution after treatment. Lines of evidence from the results of allogeneic bone marrow transplantation in patients with XSCID support this hypothesis that, however, has not been rigorously tested in an experimental system. We studied the competition kinetics between normal and XSCID bone marrow (BM) cells using a murine bone marrow transplantation (BMT) model. For easy chimerism determination, we used genetic marking with retrovirus-mediated expression of the enhanced green fluorescent protein (EGFP). We found that XSCID BM cells were able to compete with normal BM cells for engraftment of myeloid lineages in a dose-dependent manner, whereas we observed selective repopulation of T, B, and NK cells deriving from normal BM cells. This was true despite the evidence of competitive engraftment of XSCID lineage marker-negative/c-Kit-positive (Lin-/c-Kit+) cells in the bone marrow of treated animals. From these results we extrapolate that genetic correction of XSCID HSCs will result in selective advantage of gene-corrected lymphoid lineages with consequent restoration of lymphocyte populations and high probability of clinical benefit.


Subject(s)
Bone Marrow Cells/metabolism , Gene Transfer Techniques , Genetic Linkage , Genetic Therapy/methods , Severe Combined Immunodeficiency/therapy , X Chromosome/genetics , Animals , Bone Marrow Transplantation , Cell Separation , Dose-Response Relationship, Drug , Flow Cytometry , Green Fluorescent Proteins , Humans , Kinetics , Leukocytes, Mononuclear/metabolism , Luminescent Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Retroviridae/genetics
17.
Immunobiology ; 202(2): 106-19, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10993286

ABSTRACT

Combined immune deficiencies comprise a spectrum of genetic disorders characterized by developmental or functional defects of both T and B lymphocytes. Recent progress in cell biology and molecular genetics has unraveled the pathophysiology of most of these defects. In particular, the most common form of severe combined immune deficiency in humans, with lack of circulating T cells, a normal or increased number of B lymphocytes, and an X-linked pattern of inheritance (SCIDXI) has been shown to be due to defects of the IL2RG gene, encoding for the common gamma chain (gammac), shared by several cytokine receptors. Furthermore, defects of the JAK3 gene, encoding for an intracellular tyrosine kinase required for signal transduction through gammac-containing cytokine receptors, have been identified in patients with autosomal recessive T-B+ SCID. Characterization of the functional properties of cytokines that signal through the gammac-JAK3 signaling pathway has been favored by the detailed analysis of SCID patients. Specifically, the key role of IL-7 in promoting T cell development has been substantiated by the identification of rare patients with T-B+ SCID who have a defect in the alpha subunit of the IL-7 receptor (IL7Ralpha). The heterogeneity of genetic defects along the same signaling pathway that may lead to combined immune deficiency is paralleled by the heterogeneity of immunological phenotypes that may associate with defects in the same gene, thus creating a need for detailed immunological and molecular investigations in order to dissect the spectrum of combined immune deficiencies in humans.


Subject(s)
Protein-Tyrosine Kinases/immunology , Receptors, Interleukin-7/immunology , Severe Combined Immunodeficiency/immunology , Signal Transduction , Animals , Cytokines/immunology , Humans , Immunophenotyping , Interleukin Receptor Common gamma Subunit , Janus Kinase 3 , Models, Immunological , Receptors, Cytokine/immunology
18.
Hum Gene Ther ; 11(13): 1901-9, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10986562

ABSTRACT

Dendritic cells (DCs) are potent antigen-presenting cells and are capable of activating naive T cells. Gene transfer of tumor antigen and cytokine genes into DCs could be an important strategy for immunotherapeutic applications. Dendritic cells derived from peripheral blood monocytes do not divide and are therefore poor candidates for gene transfer by Moloney murine leukemia virus (Mo-MuLV)-based retroviral vectors. Lentiviral vectors are emerging as a powerful tool for gene delivery into dividing and nondividing cells. A three-plasmid expression system pseudotyped with the envelope from vesicular stomatitis virus (VSV-G) was used to generate lentiviral vector particles expressing enhanced green fluorescent protein (EGFP). Peripheral blood monocyte-derived DCs were cultured in the presence of GM-CSF and IL-4 and transduced with lentiviral or Mo-MuLV-based vectors expressing EGFP. FACS analysis of lentiviral vector-transduced DCs derived either from normal healthy volunteers or from melanoma patients demonstrated transduction efficiency ranging from 70 to 90% compared with 2-8% using Mo-MuLV-based vectors pseudotyped with VSV-G. Comparison of lentiviral vectors expressing EGFP driven by CMV or human PGK promoters showed similar levels of transgene expression. Lentiviral vector preparations produced in the absence of HIV accessory proteins transduced DCs at efficiencies equal to vectors produced with accessory proteins. Alu-HIV-1 LTR PCR demonstrated the genomic integration of the lentiviral vector in the transduced DCs. Transduced cells showed characteristic dendritic cell phenotype and strong allostimulatory capacity and maintained the ability to respond to activation signals such as CD40 ligand and lipopolysaccharide. These results provide evidence that lentiviral vectors are efficient tools for gene transfer and expression in monocyte-derived DCs that could be useful for immunotherapeutic applications.


Subject(s)
Dendritic Cells/physiology , Gene Transfer Techniques , Lentivirus/genetics , Monocytes/cytology , Alu Elements/genetics , Antigens, CD , Cytomegalovirus/genetics , Dendritic Cells/immunology , Dendritic Cells/virology , Green Fluorescent Proteins , HIV-1/genetics , Humans , Immunoglobulins/metabolism , Interleukin-12/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Melanoma/genetics , Melanoma/pathology , Membrane Glycoproteins/metabolism , Moloney murine leukemia virus/genetics , Monocytes/virology , Promoter Regions, Genetic , T-Lymphocytes/immunology , Vesicular stomatitis Indiana virus/genetics , Viral Proteins/genetics , CD83 Antigen
19.
Hum Genet ; 106(1): 73-9, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10982185

ABSTRACT

JAK3 deficiency in humans results in autosomal recessive T-B+ severe combined immunodeficiency disease (SCID), a severe immunodeficiency that can only be cured by bone marrow transplantation. We unraveled the complete organization of the human JAK3 gene, which includes 23 exons. This information allowed us to set up a molecular screening test that enabled us to diagnose JAK3 deficiency in 14 patients from 12 unrelated families with T-B+ SCID. In order to define the mutations, we used a nonradioactive single-strand conformation polymorphism (SSCP)/heteroduplex (HD) assay based on exon-specific polymerase chain reaction (PCR). In this cohort of patients, 15 independent JAK3 gene mutations have been identified, including 7 that have not been described previously. Mutation analysis information was used for genetic counseling and prenatal diagnosis.


Subject(s)
DNA Mutational Analysis/methods , Protein-Tyrosine Kinases/genetics , Severe Combined Immunodeficiency/genetics , Consanguinity , Exons , Female , Heterozygote , Homozygote , Humans , Introns , Janus Kinase 3 , Male , Models, Genetic , Molecular Sequence Data , Polymorphism, Single-Stranded Conformational , Protein-Tyrosine Kinases/deficiency
20.
Blood ; 96(4): 1309-16, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10942372

ABSTRACT

Resting lymphocytes are refractory to gene transfer using Moloney murine leukemia virus (MMLV)-based retroviral vectors because of their quiescent status. Recently, it has been shown that lentiviral vectors are capable of transferring genes into nondividing and terminally differentiated cells. We used human immunodeficiency virus type-1 (HIV-1)-based vectors expressing enhanced green fluorescent protein (EGFP) driven by different promoters (CMV, MPSV, or PGK) and investigated their ability to transduce human T- and B-cell lines, as well as resting or activated primary peripheral and umbilical cord blood lymphocytes. The effects of the presence or the absence of HIV-1 accessory proteins (Vif, Vpr, Vpu, and Nef) in the vector system were also assessed. Flow cytometry analysis showed no differences in the ability of these vectors of transferring the reporter gene into lymphocytic lines and mitogen-stimulated primary lymphocytes in the presence or the absence of HIV-1 accessory proteins (APs). Similarly, viral supernatants generated in the presence of accessory genes could efficiently transduce various subsets of resting lymphocytes and provide long-term expression of the transgene. No significant transduction-induced changes in cell activation or cycling status were observed and Alu-HIV-1 long terminal repeat polymerase chain reaction (LTR PCR) analysis demonstrated integration of the vector sequences at the molecular level. In contrast, in the absence of HIV-1 APs, lentiviral vectors failed to integrate and express the transgene in resting lymphocytes. These results show that transduction of primary resting lymphocytes with HIV-1-based vectors requires the presence of viral accessory proteins. (Blood. 2000;96:1309-1316)


Subject(s)
B-Lymphocytes , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors , HIV-1/genetics , Lentivirus , T-Lymphocytes , Cell Line , Gene Expression , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Lymphocyte Activation , Promoter Regions, Genetic , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...