Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Blood ; 137(17): 2326-2336, 2021 04 29.
Article in English | MEDLINE | ID: mdl-33545713

ABSTRACT

Immunodysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome is caused by mutations in forkhead box P3 (FOXP3), which lead to the loss of function of regulatory T cells (Tregs) and the development of autoimmune manifestations early in life. The selective induction of a Treg program in autologous CD4+ T cells by FOXP3 gene transfer is a promising approach for curing IPEX. We have established a novel in vivo assay of Treg functionality, based on adoptive transfer of these cells into scurfy mice (an animal model of IPEX) and a combination of cyclophosphamide (Cy) conditioning and interleukin-2 (IL-2) treatment. This model highlighted the possibility of rescuing scurfy disease after the latter's onset. By using this in vivo model and an optimized lentiviral vector expressing human Foxp3 and, as a reporter, a truncated form of the low-affinity nerve growth factor receptor (ΔLNGFR), we demonstrated that the adoptive transfer of FOXP3-transduced scurfy CD4+ T cells enabled the long-term rescue of scurfy autoimmune disease. The efficiency was similar to that seen with wild-type Tregs. After in vivo expansion, the converted CD4FOXP3 cells recapitulated the transcriptomic core signature for Tregs. These findings demonstrate that FOXP3 expression converts CD4+ T cells into functional Tregs capable of controlling severe autoimmune disease.


Subject(s)
Autoimmune Diseases/prevention & control , CD4-Positive T-Lymphocytes/immunology , Cyclophosphamide/pharmacology , Forkhead Transcription Factors/genetics , Genetic Diseases, X-Linked/prevention & control , Interleukin-2/pharmacology , T-Lymphocytes, Regulatory/immunology , Animals , Antineoplastic Agents/pharmacology , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , CD4-Positive T-Lymphocytes/drug effects , Disease Models, Animal , Drug Therapy, Combination , Female , Genetic Diseases, X-Linked/immunology , Genetic Diseases, X-Linked/pathology , Immunosuppressive Agents/pharmacology , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/drug effects
2.
Haematologica ; 106(2): 404-411, 2021 02 01.
Article in English | MEDLINE | ID: mdl-31919089

ABSTRACT

Severe combined immunodeficiencies (SCIDs) constitute a heterogeneous group of life-threatening genetic disorders that typically present in the first year of life. They are defined by the absence of autologous T cells and the presence of an intrinsic or extrinsic defect in the B-cell compartment. In three newborns presenting with frequent infections and profound leukopenia, we identified a private, heterozygous mutation in the RAC2 gene (p.G12R). This mutation was de novo in the index case, who had been cured by hematopoietic stem cell transplantation but had transmitted the mutation to her sick daughter. Biochemical assays showed that the mutation was associated with a gain of function. The results of in vitro differentiation assays showed that RAC2 is essential for the survival and differentiation of hematopoietic stem/progenitor cells. Therefore, screening for RAC2 gain-of-function mutations should be considered in patients with a SCID phenotype and who lack a molecular diagnosis.


Subject(s)
Hematopoietic Stem Cell Transplantation , Severe Combined Immunodeficiency , rac GTP-Binding Proteins , Bone Marrow , Bone Marrow Failure Disorders , Female , Gain of Function Mutation , Humans , Infant, Newborn , Mutation , Severe Combined Immunodeficiency/diagnosis , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/therapy , RAC2 GTP-Binding Protein
3.
Nat Genet ; 51(1): 196, 2019 01.
Article in English | MEDLINE | ID: mdl-30429576

ABSTRACT

In the version of this article originally published, the main-text sentence "In three patients of European ancestry, we identified the germline variant encoding p.Ile97Met in TIM-3, which was homozygous in two (P12 and P13) and heterozygous in one (P15) in the germline but with no TIM-3 plasma membrane expression in the tumor" misstated the identifiers of the two homozygous individuals, which should have been P13 and P14. The error has been corrected in the HTML, PDF and print versions of the paper.

4.
Nat Genet ; 50(12): 1650-1657, 2018 12.
Article in English | MEDLINE | ID: mdl-30374066

ABSTRACT

Subcutaneous panniculitis-like T cell lymphoma (SPTCL), a non-Hodgkin lymphoma, can be associated with hemophagocytic lymphohistiocytosis (HLH), a life-threatening immune activation that adversely affects survival1,2. T cell immunoglobulin mucin 3 (TIM-3) is a modulator of immune responses expressed on subgroups of T and innate immune cells. We identify in ~60% of SPTCL cases germline, loss-of-function, missense variants altering highly conserved residues of TIM-3, c.245A>G (p.Tyr82Cys) and c.291A>G (p.Ile97Met), each with specific geographic distribution. The variant encoding p.Tyr82Cys TIM-3 occurs on a potential founder chromosome in patients with East Asian and Polynesian ancestry, while p.Ile97Met TIM-3 occurs in patients with European ancestry. Both variants induce protein misfolding and abrogate TIM-3's plasma membrane expression, leading to persistent immune activation and increased production of inflammatory cytokines, including tumor necrosis factor-α and interleukin-1ß, promoting HLH and SPTCL. Our findings highlight HLH-SPTCL as a new genetic entity and identify mutations causing TIM-3 alterations as a causative genetic defect in SPTCL. While HLH-SPTCL patients with mutant TIM-3 benefit from immunomodulation, therapeutic repression of the TIM-3 checkpoint may have adverse consequences.


Subject(s)
Germ-Line Mutation , Hepatitis A Virus Cellular Receptor 2/genetics , Lymphohistiocytosis, Hemophagocytic/genetics , Lymphoma, T-Cell/genetics , Panniculitis/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Child , Diagnosis, Differential , Female , Genetic Predisposition to Disease , Humans , Infant , Lymphohistiocytosis, Hemophagocytic/classification , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphoma, T-Cell/classification , Lymphoma, T-Cell/diagnosis , Male , Middle Aged , Panniculitis/classification , Panniculitis/diagnosis , Pedigree , Exome Sequencing , Young Adult
5.
Am J Hum Genet ; 101(6): 995-1005, 2017 Dec 07.
Article in English | MEDLINE | ID: mdl-29198722

ABSTRACT

A recurrent de novo missense variant within the C-terminal Sin3-like domain of ZSWIM6 was previously reported to cause acromelic frontonasal dysostosis (AFND), an autosomal-dominant severe frontonasal and limb malformation syndrome, associated with neurocognitive and motor delay, via a proposed gain-of-function effect. We present detailed phenotypic information on seven unrelated individuals with a recurrent de novo nonsense variant (c.2737C>T [p.Arg913Ter]) in the penultimate exon of ZSWIM6 who have severe-profound intellectual disability and additional central and peripheral nervous system symptoms but an absence of frontonasal or limb malformations. We show that the c.2737C>T variant does not trigger nonsense-mediated decay of the ZSWIM6 mRNA in affected individual-derived cells. This finding supports the existence of a truncated ZSWIM6 protein lacking the Sin3-like domain, which could have a dominant-negative effect. This study builds support for a key role for ZSWIM6 in neuronal development and function, in addition to its putative roles in limb and craniofacial development, and provides a striking example of different variants in the same gene leading to distinct phenotypes.


Subject(s)
DNA-Binding Proteins/genetics , Intellectual Disability/genetics , Neurocognitive Disorders/genetics , Central Nervous System/abnormalities , Central Nervous System/embryology , Codon, Nonsense/genetics , High-Throughput Nucleotide Sequencing , Humans , Limb Deformities, Congenital/genetics , Mandibulofacial Dysostosis/genetics , Peripheral Nervous System/abnormalities , Peripheral Nervous System/enzymology
6.
Blood ; 128(1): 60-71, 2016 07 07.
Article in English | MEDLINE | ID: mdl-27222478

ABSTRACT

Hemophagocytic lymphohistiocytosis (HLH) is a life-threatening syndrome, characterized by severe hyperinflammation and immunopathological manifestations in several tissues. These features result from organ infiltration by overactivated CD8 T-cells and macrophages, which produce high levels of pro-inflammatory cytokines, such as IFN-γ, TNF-α, IL-6, and IL-18. Recently, several Janus kinase 1/2 (JAK1/2) inhibitors, such as ruxolitinib, have been developed as immunosuppressive agents. They have proven beneficial effects in the treatment of myeloproliferative disorders and inflammatory conditions. To determine whether pharmacological inhibition of the JAK1/2 not only prevents the onset of HLH immunopathology but also is effective against existing HLH, cytotoxicity-impaired Prf1(-/-) and Rab27a(-/-) mice with full-blown HLH syndrome were treated with a clinically relevant dose of ruxolitinib. In vivo, ruxolitinib treatment suppressed signal transducer and activator of transcription 1 activation and led to recovery from HLH manifestations in both murine models. In the Prf1(-/-) mice, these beneficial effects were evidenced by a greater survival rate, and in both murine models, they were evidenced by the correction of blood cytopenia and a rapid decrease in serum IL-6 and TNF-α levels. During ruxolitinib treatment, liver tissue damage receded concomitantly with a decrease in the number of infiltrating inflammatory macrophages and an increase in the number of alternatively activated macrophages. In Rab27a(-/-) mice, central nervous system involvement was significantly reduced by ruxolitinib therapy. Our findings demonstrate that clinically relevant doses of the JAK1/2 inhibitor ruxolitinib suppresses the harmful consequences of macrophage overactivation characterizing HLH in 2 murine models. The results could be readily translated into the clinic for the treatment of primary, and perhaps even secondary, forms of HLH.


Subject(s)
Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Lymphohistiocytosis, Hemophagocytic/drug therapy , Lymphohistiocytosis, Hemophagocytic/enzymology , Pyrazoles/pharmacology , Animals , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Liver/enzymology , Lymphohistiocytosis, Hemophagocytic/genetics , Macrophages/enzymology , Mice , Mice, Knockout , Nitriles , Perforin/genetics , Perforin/metabolism , Pyrimidines , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab27 GTP-Binding Proteins
7.
Blood ; 127(17): 2113-21, 2016 04 28.
Article in English | MEDLINE | ID: mdl-26864340

ABSTRACT

Hemophagocytic lymphohistiocytosis (HLH) is a life-threatening hyperinflammatory disease. Inherited forms of HLH are caused by biallelic mutations in several effectors of granule-dependent lymphocyte-mediated cytotoxicity. A small proportion of patients with a so-called "secondary" form of HLH, which develops in the aftermath of infection, autoimmunity, or cancer, carry a monoallelic mutation in one or more HLH-associated genes. Although this observation suggests that HLH may have a polygenic mode of inheritance, the latter is very difficult to prove in humans. In order to determine whether the accumulation of partial genetic defects in lymphocyte-mediated cytotoxicity can contribute to the development of HLH, we generated mice that were doubly or triply heterozygous for mutations in HLH-associated genes, those coding for perforin, Rab27a, and syntaxin-11. We found that the accumulation of monoallelic mutations did indeed increase the risk of developing HLH immunopathology after lymphocytic choriomeningitis virus infection. In mechanistic terms, the accumulation of heterozygous mutations in the two degranulation genes Rab27a and syntaxin-11, impaired the dynamics and secretion of cytotoxic granules at the immune synapse of T lymphocytes. In addition, the accumulation of heterozygous mutations within the three genes impaired natural killer lymphocyte cytotoxicity in vivo. The genetic defects can be ranked in terms of the severity of the resulting HLH manifestations. Our results form the basis of a polygenic model of the occurrence of secondary HLH.


Subject(s)
Lymphohistiocytosis, Hemophagocytic/genetics , Multifactorial Inheritance , Pore Forming Cytotoxic Proteins/genetics , Qa-SNARE Proteins/genetics , rab GTP-Binding Proteins/genetics , Animals , Cell Degranulation/genetics , Crosses, Genetic , Cytotoxicity, Immunologic/genetics , Gene Dosage , Genetic Predisposition to Disease , Heterozygote , Immunological Synapses/immunology , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Mutation , Pore Forming Cytotoxic Proteins/physiology , Qa-SNARE Proteins/physiology , Specific Pathogen-Free Organisms , T-Lymphocytes, Cytotoxic/immunology , rab GTP-Binding Proteins/physiology , rab27 GTP-Binding Proteins
8.
Blood ; 125(9): 1427-34, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25525117

ABSTRACT

The impairment of cytotoxic activity of lymphocytes disturbs immune surveillance and leads to the development of hemophagocytic lymphohistiocytic syndrome (HLH). Although cytotoxic T lymphocyte (CTL) control of HLH development is well documented, the role for natural killer (NK)-cell effector functions in the pathogenesis of this immune disorder remains unclear. In this study, we specifically targeted a defect in cytotoxicity to either CTL or NK cells in mice so as to dissect the contribution of these lymphocyte subsets to HLH-like disease severity after lymphocytic choriomeningitis virus (LCMV) infection. We found that NK-cell cytotoxicity was sufficient to protect mice from the fatal outcome that characterizes HLH-like disease and was also sufficient to reduce HLH-like manifestations. Mechanistically, NK-cell cytotoxicity reduced tissue infiltration by inflammatory macrophages and downmodulated LCMV-specific T-cell responses by limiting hyperactivation of CTL. Interestingly, the critical protective effect of NK cells on HLH was independent of interferon-γ secretion and changes in viral load. Therefore our findings identify a crucial role of NK-cell cytotoxicity in limiting HLH-like immunopathology, highlighting the important role of NK cytotoxic activity in immune homeostasis.


Subject(s)
Cytotoxicity, Immunologic/immunology , Killer Cells, Natural/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Lymphohistiocytosis, Hemophagocytic/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Proliferation , Cells, Cultured , Interferon-gamma/metabolism , Killer Cells, Natural/pathology , Killer Cells, Natural/virology , Lymphocytic Choriomeningitis/pathology , Lymphocytic Choriomeningitis/virology , Lymphohistiocytosis, Hemophagocytic/pathology , Lymphohistiocytosis, Hemophagocytic/virology , Mice , Mice, Inbred C57BL , Spleen/immunology , Spleen/pathology , Spleen/virology , Viral Load
9.
PLoS One ; 9(8): e102902, 2014.
Article in English | MEDLINE | ID: mdl-25089524

ABSTRACT

The tumor suppressor retinoblastoma protein (pRb) is inactivated in a wide variety of cancers. While its role during cell cycle is well characterized, little is known about its properties on apoptosis regulation and apoptosis-induced cell responses. pRb shorter forms that can modulate pRB apoptotic properties, resulting from cleavages at caspase specific sites are observed in several cellular contexts. A bioinformatics analysis showed that a putative caspase cleavage site (TELD) is found in the Drosophila homologue of pRb(RBF) at a position similar to the site generating the p76Rb form in mammals. Thus, we generated a punctual mutant form of RBF in which the aspartate of the TELD site is replaced by an alanine. This mutant form, RBFD253A, conserved the JNK-dependent pro-apoptotic properties of RBF but gained the ability of inducing overgrowth phenotypes in adult wings. We show that this overgrowth is a consequence of an abnormal proliferation in wing imaginal discs, which depends on the JNK pathway activation but not on wingless (wg) ectopic expression. These results show for the first time that the TELD site of RBF could be important to control the function of RBF in tissue homeostasis in vivo.


Subject(s)
Apoptosis , Drosophila Proteins/genetics , Homeostasis , Mutation/genetics , Organ Specificity , Retinoblastoma Protein/genetics , Transcription Factors/genetics , Aging , Animals , Caspases/metabolism , Cell Proliferation , Consensus Sequence , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Imaginal Discs/cytology , Imaginal Discs/growth & development , Imaginal Discs/metabolism , Larva/cytology , Larva/metabolism , MAP Kinase Signaling System , Phenotype , Retinoblastoma Protein/metabolism , Transcription Factors/metabolism , Wings, Animal/cytology , Wings, Animal/growth & development , Wings, Animal/metabolism , Wnt1 Protein/metabolism
10.
Blood Cells Mol Dis ; 47(1): 72-8, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21531153

ABSTRACT

An important proof of principle has been achieved with the development of an in vitro T-cell differentiation assay based on the coculture of hematopoietic progenitors with the OP9-Delta1 stromal cell line. The original murine T cell differentiation assay has since been adapted for human T-cell differentiation, however with lower efficiency. The choice of both medium and cytokines is crucial in this assay, therefore our work has been focused on these two factors. The use of freshly reconstituted medium, the optimization of interleukine-7 (IL-7) concentration, and the addition of stem cell factor (SCF) have allowed to improve the proliferation of progenitors and T-cell precursors as well as the yield of double positive CD4+CD8+ T cells, and mature γδ and αß T cells. These optimizations make the OP9-Delta1 system sensitive enough to perform both quantitative and qualitative assays with various type of progenitors, including those transduced by a retroviral vector. The improved OP9-Delta1 assay therefore constitutes an extremely useful test for basic research purposes and for translational medicine.


Subject(s)
Cell Differentiation/drug effects , Culture Media, Conditioned/pharmacology , Cytokines/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Adjuvants, Immunologic/pharmacology , Cells, Cultured , Coculture Techniques , Cytokines/immunology , Hematopoietic Stem Cells/immunology , Humans , Interleukin-7/pharmacology , Stem Cell Factor/pharmacology , Stromal Cells/immunology
11.
J Clin Invest ; 118(9): 3132-42, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18688285

ABSTRACT

Previously, several individuals with X-linked SCID (SCID-X1) were treated by gene therapy to restore the missing IL-2 receptor gamma (IL2RG) gene to CD34+ BM precursor cells using gammaretroviral vectors. While 9 of 10 patients were successfully treated, 4 of the 9 developed T cell leukemia 31-68 months after gene therapy. In 2 of these cases, blast cells contained activating vector insertions near the LIM domain-only 2 (LMO2) proto-oncogene. Here, we report data on the 2 most recent adverse events, which occurred in patients 7 and 10. In patient 10, blast cells contained an integrated vector near LMO2 and a second integrated vector near the proto-oncogene BMI1. In patient 7, blast cells contained an integrated vector near a third proto-oncogene,CCND2. Additional genetic abnormalities in the patients' blast cells included chromosomal translocations, gain-of-function mutations activating NOTCH1, and copy number changes, including deletion of tumor suppressor gene CDKN2A, 6q interstitial losses, and SIL-TAL1 rearrangement. These findings functionally specify a genetic network that controls growth in T cell progenitors. Chemotherapy led to sustained remission in 3 of the 4 cases of T cell leukemia, but failed in the fourth. Successful chemotherapy was associated with restoration of polyclonal transduced T cell populations. As a result, the treated patients continued to benefit from therapeutic gene transfer.


Subject(s)
Chromosomes, Human, X , Genetic Therapy/adverse effects , Genetic Therapy/methods , Leukemia, T-Cell/etiology , Severe Combined Immunodeficiency/therapy , Adaptor Proteins, Signal Transducing , Antineoplastic Agents/pharmacology , Chromosome Aberrations , Cyclin D2 , Cyclins/genetics , DNA-Binding Proteins/genetics , Gammaretrovirus/metabolism , Humans , Infant , Janus Kinase 3/genetics , LIM Domain Proteins , Leukemia, T-Cell/complications , Leukemia, T-Cell/therapy , Metalloproteins/genetics , Models, Biological , Mutation , Proto-Oncogene Mas , Proto-Oncogene Proteins , Receptors, Interleukin-2/genetics , Severe Combined Immunodeficiency/complications
12.
Mol Ther ; 16(8): 1490-9, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18560421

ABSTRACT

Patients with mutations in the Artemis gene display a complete absence of T- and B lymphocytes, together with increased cellular radiosensitivity; this leads to a radiosensitive severe combined immunodeficiency (RS-SCID). Allogenic hematopoietic stem-cell (HSC) transplantation is only partially successful in the absence of an human leukocyte antigen-genoidentical donor, and this has prompted a search for alternative therapeutic approaches such as gene therapy. In this study, a self-inactivated lentiviral vector expressing Artemis was used to complement the Artemis knockout mouse (Art(-/-)). Transplantation of Artemis-transduced HSCs into irradiated Art(-/-) mice restored a stable (over a 15-month period of follow-up) and functional T- and cell repertoire that was comparable to that of control mice. The success of secondary transplantations demonstrated that the HSCs had been transduced. One of thirteen mice developed a thymoma 6 months after gene therapy. Although thymic cells were seen to be carrying two lentiviral integration sites, there was no evidence of lentivirus-driven oncogene activation. The Art(-/-) mice were found to be prone to develop T-cell lymphomas, either spontaneously or after irradiation. These data indicate that the observed lymphoproliferation was probably the consequence of the chromosomal instability associated with the Artemis-deficient background. As a whole, our work provides a basis for supporting the gene therapy approach in Artemis-deficient SCID.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Lentivirus/genetics , Nuclear Proteins/genetics , Severe Combined Immunodeficiency/therapy , Animals , B-Lymphocytes/immunology , CD3 Complex/analysis , CD4 Antigens/analysis , CD8 Antigens/analysis , Cells, Cultured , Endonucleases , Female , Flow Cytometry , Genetic Therapy/methods , Genetic Vectors/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Hyaluronan Receptors/analysis , Interleukin-2 Receptor alpha Subunit/analysis , Male , Mice , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/metabolism , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/immunology , T-Lymphocytes/immunology , Thymus Gland/immunology , Thymus Gland/pathology , Transduction, Genetic
13.
Nucleic Acids Res ; 36(9): e49, 2008 May.
Article in English | MEDLINE | ID: mdl-18411205

ABSTRACT

Gene transfer has been used to correct inherited immunodeficiencies, but in several patients integration of therapeutic retroviral vectors activated proto-oncogenes and caused leukemia. Here, we describe improved methods for characterizing integration site populations from gene transfer studies using DNA bar coding and pyrosequencing. We characterized 160,232 integration site sequences in 28 tissue samples from eight mice, where Rag1 or Artemis deficiencies were corrected by introducing the missing gene with gamma-retroviral or lentiviral vectors. The integration sites were characterized for their genomic distributions, including proximity to proto-oncogenes. Several mice harbored abnormal lymphoproliferations following therapy--in these cases, comparison of the location and frequency of isolation of integration sites across multiple tissues helped clarify the contribution of specific proviruses to the adverse events. We also took advantage of the large number of pyrosequencing reads to show that recovery of integration sites can be highly biased by the use of restriction enzyme cleavage of genomic DNA, which is a limitation in all widely used methods, but describe improved approaches that take advantage of the power of pyrosequencing to overcome this problem. The methods described here should allow integration site populations from human gene therapy to be deeply characterized with spatial and temporal resolution.


Subject(s)
Genetic Therapy/adverse effects , Sequence Analysis, DNA/methods , Animals , Cell Proliferation , DNA Restriction Enzymes , Disease Models, Animal , Gene Transfer Techniques , Genes, Tumor Suppressor , Genetic Vectors , Lentivirus/genetics , Lymphoproliferative Disorders/genetics , Mice , Proto-Oncogenes
14.
J Exp Med ; 204(13): 3085-93, 2007 Dec 24.
Article in English | MEDLINE | ID: mdl-18070935

ABSTRACT

Identification of a thymus-seeding progenitor originating from human bone marrow (BM) constitutes a key milestone in understanding the mechanisms of T cell development and provides new potential for correcting T cell deficiencies. We report the characterization of a novel lymphoid-restricted subset, which is part of the lineage-negative CD34(+)CD10(+) progenitor population and which is distinct from B cell-committed precursors (in view of the absence of CD24 expression). We demonstrate that these Lin(-)CD34(+)CD10(+)CD24(-) progenitors have a very low myeloid potential but can generate B, T, and natural killer lymphocytes and coexpress recombination activating gene 1, terminal deoxynucleotide transferase, PAX5, interleukin 7 receptor alpha, and CD3epsilon. These progenitors are present in the cord blood and in the BM but can also be found in the blood throughout life. Moreover, they belong to the most immature thymocyte population. Collectively, these findings unravel the existence of a postnatal lymphoid-polarized population that is capable of migrating from the BM to the thymus.


Subject(s)
Stem Cells/metabolism , Thymus Extracts/metabolism , Thymus Gland/metabolism , Antigens, CD34/biosynthesis , Bone Marrow Cells/metabolism , CD24 Antigen/biosynthesis , CD3 Complex/biosynthesis , Cells, Cultured , Flow Cytometry , Gene Expression Profiling , Humans , Leukocytes, Mononuclear/metabolism , Neprilysin/biosynthesis , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology
15.
J Clin Invest ; 117(8): 2225-32, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17671652

ABSTRACT

Recent reports have challenged the notion that retroviruses and retroviral vectors integrate randomly into the host genome. These reports pointed to a strong bias toward integration in and near gene coding regions and, for gammaretroviral vectors, around transcription start sites. Here, we report the results obtained from a large-scale mapping of 572 retroviral integration sites (RISs) isolated from cells of 9 patients with X-linked SCID (SCID-X1) treated with a retrovirus-based gene therapy protocol. Our data showed that two-thirds of insertions occurred in or very near to genes, of which more than half were highly expressed in CD34(+) progenitor cells. Strikingly, one-fourth of all integrations were clustered as common integration sites (CISs). The highly significant incidence of CISs in circulating T cells and the nature of their locations indicate that insertion in many gene loci has an influence on cell engraftment, survival, and proliferation. Beyond the observed cases of insertional mutagenesis in 3 patients, these data help to elucidate the relationship between vector insertion and long-term in vivo selection of transduced cells in human patients with SCID-X1.


Subject(s)
Gammaretrovirus , Genetic Therapy , Genetic Vectors , Genome, Human , Lymphopoiesis/genetics , Virus Integration/genetics , X-Linked Combined Immunodeficiency Diseases/therapy , Antigens, CD34 , Cell Proliferation , Cell Survival/genetics , Hematopoietic Stem Cells/metabolism , Humans , Mutagenesis, Insertional , Quantitative Trait Loci , T-Lymphocytes , Time Factors , X-Linked Combined Immunodeficiency Diseases/genetics
16.
PLoS One ; 2(6): e570, 2007 Jun 27.
Article in English | MEDLINE | ID: mdl-17593969

ABSTRACT

Features such as mutations or structural characteristics can be non-randomly or non-uniformly distributed within a genome. So far, computer simulations were required for statistical inferences on the distribution of sequence motifs. Here, we show that these analyses are possible using an analytical, mathematical approach. For the assessment of non-randomness, our calculations only require information including genome size, number of (sampled) sequence motifs and distance parameters. We have developed computer programs evaluating our analytical formulas for the real-time determination of expected values and p-values. This approach permits a flexible cluster definition that can be applied to most effectively identify non-random or non-uniform sequence motif distribution. As an example, we show the effectivity and reliability of our mathematical approach in clinical retroviral vector integration site distribution.


Subject(s)
Genome, Human , Retroviridae/genetics , Virus Integration/genetics , Cluster Analysis , Computer Simulation , Humans
17.
Blood ; 107(1): 63-72, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16174758

ABSTRACT

Severe combined immunodeficiency (SCID) caused by mutations in RAG1 or RAG2 genes is characterized by a complete block in T- and B-cell development. The only curative treatment is allogeneic hematopoietic stem cell transplantation, which gives a high survival rate (90%) when an HLA-genoidentical donor exists but unsatisfactory results when only partially compatible donors are available. We have thus been interested in the development of a potential alternative treatment by using retroviral gene transfer of a normal copy of RAG1 cDNA. We show here that this approach applied to RAG-1-deficient mice restores normal B- and T-cell function even in the presence of a reduced number of mature B cells. The reconstitution is stable over time, attesting to a selective advantage of transduced progenitors. Notably, a high transgene copy number was detected in all lymphoid organs, and this was associated with a risk of lymphoproliferation as observed in one mouse. Altogether, these results demonstrate that correction of RAG-1 deficiency can be achieved by gene therapy in immunodeficient mice but that human application would require the use of self-inactivated vector to decrease the risk of lymphoproliferative diseases.


Subject(s)
Genetic Therapy/methods , Homeodomain Proteins/administration & dosage , Immune System/drug effects , Regeneration/drug effects , Severe Combined Immunodeficiency/therapy , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Gene Dosage , Genetic Therapy/adverse effects , Homeodomain Proteins/adverse effects , Homeodomain Proteins/genetics , Immune System/physiology , Lymphoproliferative Disorders/etiology , Mice , Mice, Knockout , Retroviridae/genetics , Severe Combined Immunodeficiency/complications , T-Lymphocytes/cytology , T-Lymphocytes/immunology
18.
Blood ; 105(7): 2699-706, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15585650

ABSTRACT

Immune function has been restored in 9 of 10 children with X-linked severe combined immunodeficiency by gamma c gene transfer in CD34+ cells. The distribution of both T-cell receptor (TCR) V beta family usage and TCR V beta complementarity-determining region 3 (CDR3) length revealed a broadly diversified T-cell repertoire. Retroviral integration site analysis in T cells demonstrated a high number of distinct insertion sites, indicating polyclonality of genetically corrected cell clones, in all patients. Detection of gamma c transgene expression on patients' mature myeloid cells has prompted us to investigate the nature of the most immature transduced hematopoietic precursor cells. Insertion sites shared by T and B lymphocytes as well as highly purified granulocytes and monocytes demonstrate the correction of common multipotent progenitor cells. Moreover, our data show that differentiated leukocytes share the same exact insertion sites with CD34+ cells that we obtained 8 months later and that were able to generate long-term culture-initiating cells (LTC-ICs). This finding demonstrates the initial transduction of very primitive multipotent progenitor cells with self-renewal capacity. These results provide a first evidence in the setting of a clinical trial that CD34+ cells maintain both lymphomyeloid potential as well as self-renewal capacity after ex vivo manipulation.


Subject(s)
Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Severe Combined Immunodeficiency/therapy , T-Lymphocytes/cytology , Antigens, CD34/immunology , Cell Differentiation/immunology , Cell Division/immunology , Cells, Cultured , Clone Cells , Gene Transfer Techniques , Granulocytes/cytology , Granulocytes/physiology , Hematopoiesis , Hematopoietic Stem Cells/immunology , Humans , Retroviridae/genetics , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/immunology , T-Lymphocytes/physiology , Transduction, Genetic
19.
Bull Acad Natl Med ; 189(5): 779-85; discussion 786-8, 2005 May.
Article in French | MEDLINE | ID: mdl-16433450

ABSTRACT

Inherited defects in T lymphocyte development (severe combined immunodeficiencies-SCID) are considered the best available models to assess the effectiveness of gene therapy. Retroviral vectors have been successfully used ex vivo to transduce hematopoietic precursors from patients with two forms of SCID, namely X-linked SCID and ADA deficiency. Fifteen out of 16 patients with XL-SCID and 4 out of 4 patients with ADA deficiency have benefited from gene therapy. Correction of the immunodeficiency has been maintained now for 6 years in the first patients to be treated. These results provide clear-cut proof of the principle of gene therapy in relevant clinical models. However, three patients with XL-SCID have developed a severe complication after gene therapy, consisting of leukemia-like clonal lymphocyte proliferation. One of the three patients has died from this complication. It is due to provirus insertion within or close to proto-oncogenes, and to the enhancer activity of the viral LTR on targeted proto-oncogenes. Vector modifications, based mostly on inactivating the enhancer activity of the LTR, should preserve efficacy while improving safety.


Subject(s)
Genetic Therapy/methods , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/therapy , Female , Genetic Therapy/adverse effects , Genetic Vectors/therapeutic use , Hematopoietic Stem Cell Transplantation , Humans , Infant , Infant, Newborn , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL