Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Blood Adv ; 7(20): 6351-6363, 2023 10 24.
Article in English | MEDLINE | ID: mdl-37595058

ABSTRACT

Mutations in the transcription factor GATA2 can cause MonoMAC syndrome, a GATA2 deficiency disease characterized by several findings, including disseminated nontuberculous mycobacterial infections, severe deficiencies of monocytes, natural killer cells, and B lymphocytes, and myelodysplastic syndrome. GATA2 mutations are found in âˆ¼90% of patients with a GATA2 deficiency phenotype and are largely missense mutations in the conserved second zinc-finger domain. Mutations in an intron 5 regulatory enhancer element are also well described in GATA2 deficiency. Here, we present a multigeneration kindred with the clinical features of GATA2 deficiency but lacking an apparent GATA2 mutation. Whole genome sequencing revealed a unique adenine-to-thymine variant in the GATA2 -110 enhancer 116,855 bp upstream of the GATA2 ATG start site. The mutation creates a new E-box consensus in position with an existing GATA-box to generate a new hematopoietic regulatory composite element. The mutation segregates with the disease in several generations of the family. Cell type-specific allelic imbalance of GATA2 expression was observed in the bone marrow of a patient with higher expression from the mutant-linked allele. Allele-specific overexpression of GATA2 was observed in CRISPR/Cas9-modified HL-60 cells and in luciferase assays with the enhancer mutation. This study demonstrates overexpression of GATA2 resulting from a single nucleotide change in an upstream enhancer element in patients with MonoMAC syndrome. Patients in this study were enrolled in the National Institute of Allergy and Infectious Diseases clinical trial and the National Cancer Institute clinical trial (both trials were registered at www.clinicaltrials.gov as #NCT01905826 and #NCT01861106, respectively).


Subject(s)
GATA2 Deficiency , Myelodysplastic Syndromes , Humans , GATA2 Deficiency/genetics , Regulatory Sequences, Nucleic Acid , Myelodysplastic Syndromes/genetics , Mutation , Gene Expression Regulation , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism
2.
Blood Adv ; 6(3): 793-807, 2022 02 08.
Article in English | MEDLINE | ID: mdl-34529785

ABSTRACT

Patients with GATA2 deficiencyharbor de novo or inherited germline mutations in the GATA2 transcription factor gene, predisposing them to myeloid malignancies. There is considerable variation in disease progression, even among family members with the same mutation in GATA2. We investigated somatic mutations in 106 patients with GATA2 deficiency to identify acquired mutations that are associated with myeloid malignancies. Myelodysplastic syndrome (MDS) was the most common diagnosis (∼44%), followed by GATA2 bone marrow immunodeficiency disorder (G2BMID; ∼37%). Thirteen percent of the cohort had GATA2 mutations but displayed no disease manifestations. There were no correlations between age or sex with disease progression or survival. Cytogenetic analyses showed a high incidence of abnormalities (∼43%), notably trisomy 8 (∼23%) and monosomy 7 (∼12%), but the changes did not correlate with lower survival. Somatic mutations in ASXL1 and STAG2 were detected in ∼25% of patients, although the mutations were rarely concomitant. Mutations in DNMT3A were found in ∼10% of patients. These somatic mutations were found similarly in G2BMID and MDS, suggesting clonal hematopoiesis in early stages of disease, before the onset of MDS. ASXL1 mutations conferred a lower survival probability and were more prevalent in female patients. STAG2 mutations also conferred a lower survival probability, but did not show a statistically significant sex bias. There was a conspicuous absence of many commonly mutated genes associated with myeloid malignancies, including TET2, IDH1/2, and the splicing factor genes. Notably, somatic mutations in chromatin-related genes and cohesin genes characterized disease progression in GATA2 deficiency.


Subject(s)
GATA2 Deficiency , Myelodysplastic Syndromes , Myeloproliferative Disorders , Neoplasms , Cell Cycle Proteins/genetics , Disease Progression , Female , GATA2 Deficiency/complications , GATA2 Deficiency/genetics , GATA2 Transcription Factor/genetics , Humans , Mutation , Myelodysplastic Syndromes/pathology , Repressor Proteins/genetics
3.
Mol Ther ; 21(5): 964-72, 2013 May.
Article in English | MEDLINE | ID: mdl-23531552

ABSTRACT

The development of leukemia following gammaretroviral vector-mediated gene therapy for X-linked severe combined immunodeficiency disease and chronic granulomatous disease (CGD) has emphasized the need for long-term follow-up in animals treated with hematopoietic stem cell gene therapy. In this study, we report the long-term follow-up (4-7 years) of four dogs with canine leukocyte adhesion deficiency (CLAD) treated with foamy viral (FV) vector-mediated gene therapy. All four CLAD dogs previously received nonmyeloablative conditioning with 200 cGy total body irradiation followed by infusion of autologous, CD34(+) hematopoietic stem cells transduced by a FV vector expressing canine CD18 from an internal Murine Stem Cell Virus (MSCV) promoter. CD18(+) leukocyte levels were >2% following infusion of vector-transduced cells leading to ongoing reversal of the CLAD phenotype for >4 years. There was no clinical development of lymphoid or myeloid leukemia in any of the four dogs and integration site analysis did not reveal insertional oncogenesis. These results showing disease correction/amelioration of disease in CLAD without significant adverse events provide support for the use of a FV vector to treat children with leukocyte adhesion deficiency type 1 (LAD-1) in a human gene therapy clinical trial.


Subject(s)
Genetic Therapy , Genetic Vectors/genetics , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocyte-Adhesion Deficiency Syndrome/therapy , Spumavirus/genetics , Animals , Antigens, CD34/metabolism , Bone Marrow , CD18 Antigens/metabolism , Disease Models, Animal , Dogs , Female , Follow-Up Studies , Gene Transfer Techniques , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Humans , Leukocyte Count , Leukocytes/metabolism , Male , T-Lymphocyte Subsets/metabolism , Transduction, Genetic , Virus Integration
4.
Hum Gene Ther ; 22(6): 689-96, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21275758

ABSTRACT

Children with leukocyte adhesion deficiency type 1 (LAD-1) and dogs with canine LAD (CLAD) develop life-threatening bacterial infections due to mutations in the leukocyte integrin CD18. Here, we compared the human phosphoglycerate kinase (hPGK) promoter to the murine stem cell virus (MSCV) promoter/enhancer in a self-inactivating HIV-1-derived lentiviral vector to treat animals with CLAD. Four CLAD dogs were infused with CD34(+) cells transduced with the hPGK vector, and two CLAD dogs received MSCV vector-transduced CD34(+) cells. Infusions were preceded by a nonmyeloablative dose of 200 cGy total body irradiation. Comparable numbers of transduced cells were infused in each group of animals. Only one of four CLAD animals treated with the hPGK-cCD18 vector had reversal of CLAD, whereas both MSCV-cCD18 vector-treated dogs had reversal of the phenotype. Correction of CLAD depends both upon the percentage of CD18(+) myeloid cells and the level of expression of CD18 on individual myeloid cells. In this regard, the hPGK promoter directed low levels of expression of CD18 on neutrophils compared to the MSCV promoter, likely contributing to the suboptimal clinical outcome with the hPGK vector.


Subject(s)
CD18 Antigens/genetics , Dog Diseases/therapy , Genetic Therapy/methods , Leukocyte-Adhesion Deficiency Syndrome/therapy , Leukocyte-Adhesion Deficiency Syndrome/veterinary , Animals , Dogs , Genetic Vectors , HIV-1/genetics , Humans , Lentivirus/genetics , Mice , Phosphoglycerate Kinase/genetics , Promoter Regions, Genetic , Stem Cells , Transduction, Genetic/methods , Whole-Body Irradiation
5.
Mol Ther ; 19(1): 113-21, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20859258

ABSTRACT

To identify cellular promoters in a self-inactivating (SIN) lentiviral vector that might be beneficial in treating children with leukocyte adhesion deficiency type 1 (LAD-1), we tested lentiviral vectors with human CD11 and CD18 leukocyte integrin proximal promoter elements directing expression of canine CD18 in animals with canine LAD (CLAD). Lentiviral vectors with either the human CD11b (637 bp) proximal promoter or the human CD18 (1,060 bp) proximal promoter resulted in the highest percentages of CD18(+) CLAD CD34(+) cells in vitro. Subsequently, two CLAD dogs were infused with autologous CD34(+) cells transduced with the hCD11b (637 bp)-cCD18 vector, and two CLAD dogs were infused with autologous CD34(+) cells transduced with the hCD18 (1,060 bp)-cCD18 vector. Each dog received a nonmyeloablative dose of 200 cGy total body irradiation (TBI) before the infusion of transduced cells. The two CLAD dogs treated with the hCD18 (1,060 bp)-cCD18 vector, and one of the two dogs treated with the hCD11b (637 bp)-cCD18 vector, had reversal of the CLAD phenotype. These studies using endogenous leukocyte integrin proximal promoters represent an important step in the development of gene therapy for children with LAD-1.


Subject(s)
CD11b Antigen/genetics , CD18 Antigens/genetics , Genetic Therapy/methods , Lentivirus/genetics , Animals , Antigens, CD34/genetics , CD11b Antigen/biosynthesis , CD18 Antigens/biosynthesis , Dogs , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocyte-Adhesion Deficiency Syndrome/therapy , Neutrophils/immunology , Neutrophils/metabolism , Promoter Regions, Genetic , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transduction, Genetic/methods , Treatment Outcome , Whole-Body Irradiation/methods
6.
Biol Blood Marrow Transplant ; 15(3): 293-305, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19203720

ABSTRACT

In the murine model, in utero hematopoietic cell transplantation (IUHCT) has been shown to achieve low levels of allogeneic chimerism and associated donor-specific tolerance permitting minimal conditioning postnatal hematopoietic stem cell transplantation (HSCT). In this pilot study, we investigated IUHCT in the canine leukocyte adhesion deficiency (CLAD) model. Haploidentical IUHCT resulted in stable low-level donor cell chimerism in all dogs that could be analyzed by sensitive detection methodology (4 of 10) through 18 months of follow-up. In the 2 CLAD recipients, low-level chimerism resulted in amelioration and complete reversal of the CLAD phenotype, respectively. Six recipients of IUHCT (5 carriers and 1 CLAD) subsequently received postnatal HSCT from the same haploidentical prenatal donor after minimal conditioning with busulfan 10 mg/kg. Chimerism in 2 of 5 CLAD carriers that underwent HSCT increased from < 1% pre-HSCT to sustained levels of 35% to 45%. Control animals undergoing postnatal haploidentical HSCT without IUHCT had no detectable donor chimerism. These results demonstrate that haploidentical IUHCT in the CLAD model can result in low-level donor chimerism that can prevent the lethal phenotype in CLAD dogs, and can result in donor-specific tolerance that can facilitate postnatal minimal conditioning HSCT.


Subject(s)
Fetal Therapies/methods , Hematopoietic Stem Cell Transplantation/methods , Leukocyte-Adhesion Deficiency Syndrome/therapy , Animals , Busulfan/administration & dosage , Dipeptidyl-Peptidase IV Inhibitors , Disease Models, Animal , Dogs , Female , Graft vs Host Disease/immunology , Haploidy , Immune Tolerance , Immunosuppressive Agents/administration & dosage , Leukocyte-Adhesion Deficiency Syndrome/immunology , Lymphocyte Culture Test, Mixed , Male , Mice , Pregnancy , Transplantation Chimera , Transplantation Conditioning/methods
7.
Nat Med ; 14(1): 93-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18157138

ABSTRACT

Recent successes in treating genetic immunodeficiencies have demonstrated the therapeutic potential of stem cell gene therapy. However, the use of gammaretroviral vectors in these trials led to insertional activation of nearby oncogenes and leukemias in some study subjects, prompting studies of modified or alternative vector systems. Here we describe the use of foamy virus vectors to treat canine leukocyte adhesion deficiency (CLAD). Four of five dogs with CLAD that received nonmyeloablative conditioning and infusion of autologous, CD34+ hematopoietic stem cells transduced by a foamy virus vector expressing canine CD18 had complete reversal of the CLAD phenotype, which was sustained more than 2 years after infusion. In vitro assays showed correction of the lymphocyte proliferation and neutrophil adhesion defects that characterize CLAD. There were no genotoxic complications, and integration site analysis showed polyclonality of transduced cells and a decreased risk of integration near oncogenes as compared to gammaretroviral vectors. These results represent the first successful use of a foamy virus vector to treat a genetic disease, to our knowledge, and suggest that foamy virus vectors will be effective in treating human hematopoietic diseases.


Subject(s)
Genetic Therapy/methods , Genetic Vectors , Leukocyte-Adhesion Deficiency Syndrome/therapy , Leukocytes/cytology , Spumavirus/genetics , Animals , Antigens, CD34/biosynthesis , Bone Marrow Cells/metabolism , Cell Adhesion , Cell Proliferation , Dogs , Hematopoietic Stem Cells/metabolism , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocyte-Adhesion Deficiency Syndrome/veterinary , Lymphocytes/metabolism , Phenotype
8.
Blood ; 108(10): 3313-20, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-16868255

ABSTRACT

Canine leukocyte adhesion deficiency (CLAD) represents the canine counter-part of the human disease leukocyte adhesion deficiency (LAD). Defects in the leukocyte integrin CD18 adhesion molecule in both CLAD and LAD lead to recurrent, life-threatening bacterial infections. We evaluated ex vivo retroviral-mediated gene therapy in CLAD using 2 nonmyeloablative conditioning regimens--200 cGy total body irradiation (TBI) or 10 mg/kg busulfan--with or without posttransplantation immunosuppression. In 6 of 11 treated CLAD dogs, therapeutic levels of CD18(+) leukocytes were achieved. Conditioning with either TBI or busulfan allowed long-term engraftment, and immunosuppression was not required for efficacy. The percentage of CD18(+) leukocytes in the peripheral blood progressively increased over 6 to 8 months after infusion to levels ranging from 1.26% to 8.37% at 1-year follow-up in the 6 dogs. These levels resulted in reversal or moderation of the severe CLAD phenotype. Linear amplification-mediated polymerase chain reaction assays indicated polyclonality of insertion sites. These results describe ex vivo hematopoietic stem cell gene transfer in a disease-specific, large animal model using 2 clinically applicable conditioning regimens, and they provide support for the use of nonmyeloablative conditioning regimens in preclinical protocols of retroviral-mediated gene transfer for nonmalignant hematopoietic diseases such as LAD.


Subject(s)
Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation/methods , Leukocyte-Adhesion Deficiency Syndrome/therapy , Transplantation Conditioning/methods , Animals , CD18 Antigens/genetics , Dogs , Genetic Vectors , Hematopoietic Stem Cells/metabolism , Retroviridae , Transfection , Whole-Body Irradiation
9.
Blood ; 108(5): 1767-9, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16645166

ABSTRACT

Nonmyeloablative conditioning regimens are increasingly replacing myeolablative conditioning prior to allogeneic hematopoietic stem cell transplantation (SCT). The recent advent of these conditioning regimens has limited the assessment of the long-term effects of this treatment, including analysis of reproductive function. To address the question of reproductive function after nonmyeloablative transplantation, we analyzed a cohort of young dogs with the genetic disease canine leukocyte adhesion deficiency that were treated with a nonmyeloablative dose of 200 cGy total body irradiation followed by matched-littermate SCT. Five males and 5 females entered puberty; all 5 males and 4 females subsequently sired or delivered litters following transplantation. We demonstrate that fertility is intact and dogs have uncomplicated parturitions following nonmyeloablative conditioning for SCT. These results are encouraging for children and adults of childbearing age who receive similar conditioning regimens prior to allogeneic transplantation.


Subject(s)
Dog Diseases/therapy , Estrus/physiology , Leukocyte-Adhesion Deficiency Syndrome/veterinary , Reproduction/physiology , Stem Cell Transplantation/veterinary , Animals , Dogs , Female , Leukocyte-Adhesion Deficiency Syndrome/therapy , Male , Pregnancy , Sperm Count , Sperm Motility , Transplantation, Homologous
10.
Biol Blood Marrow Transplant ; 11(10): 755-63, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16182176

ABSTRACT

Leukocyte adhesion deficiency (LAD)-1, a primary immunodeficiency disease caused by molecular defects in the leukocyte integrin CD18 molecule, is characterized by recurrent, life-threatening bacterial infections. Myeloablative hematopoietic stem cell transplantation is the only curative treatment for LAD-1. Recently, canine LAD (CLAD) has been shown to be a valuable animal model for the preclinical testing of nonmyeloablative transplantation regimens for the treatment of children with LAD-1. To develop new allogeneic transplantation approaches for LAD-1, we assessed a nonmyeloablative conditioning regimen consisting of busulfan as a single agent before matched littermate allogeneic bone marrow transplantation in CLAD. Three CLAD dogs received busulfan 10 mg/kg intravenously before infusion of matched littermate bone marrow, and all dogs received posttransplantation immunosuppression with cyclosporin A and mycophenolate mofetil. Initially, all 3 dogs became mixed chimeras, and levels of donor chimerism sufficient to reverse the CLAD phenotype persisted in 2 animals. The third dog maintained donor microchimerism with an attenuated CLAD phenotype. These 3 dogs have all been followed up for at least 1 year after transplantation. These results indicate that a nonmyeloablative conditioning regimen with chemotherapy alone is capable of generating stable mixed chimerism and reversal of the disease phenotype in CLAD.


Subject(s)
Bone Marrow Transplantation/methods , Busulfan/administration & dosage , Leukocyte-Adhesion Deficiency Syndrome/therapy , Transplantation Conditioning/methods , Animals , Cyclosporine/administration & dosage , Dog Diseases/therapy , Dogs , Follow-Up Studies , Immunosuppressive Agents/therapeutic use , Leukocyte-Adhesion Deficiency Syndrome/mortality , Mycophenolic Acid/administration & dosage , Mycophenolic Acid/analogs & derivatives , Phenotype , Transplantation Chimera , Treatment Outcome
11.
Exp Hematol ; 33(6): 706-12, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15911095

ABSTRACT

OBJECTIVE: The aim of this study was to test a nonmyeloablative hematopoietic stem cell transplant regimen applicable to children with leukocyte adhesion deficiency (LAD) who have a histocompatible sibling donor by using the canine model of LAD, namely canine leukocyte adhesion deficiency or CLAD. METHODS: Thirteen CLAD pups received a hematopoietic stem cell transplant from a dog leukocyte antigen (DLA)-matched littermate donor after pretransplant nonmyeloablative conditioning with 200 cGy total-body irradiation and posttransplant immunosuppression with cyclosporine and mycophenolate mofetil. Donor chimerism following transplant was assessed by flow cytometry for the presence of donor CD18 peripheral blood leukocytes and leukocyte subsets. RESULTS: Eleven of the 13 transplanted animals achieved stable mixed donor chimerism and reversal of the severe CLAD phenotype without graft-vs-host disease. The level of donor chimerism ranged from 3.9 to 95.5% at 1 year following transplant. There was one early death 3 weeks after transplant from thrombocytopenia and hemorrhage, and one dog with donor microchimerism (0.5% CD18+ donor leukocytes) who had attenuation of the CLAD phenotype. CONCLUSION: These results demonstrate that a nonmyeloablative transplant regimen from a DLA-matched littermate donor leads to mixed chimerism and reversal of the severe disease phenotype in dogs with CLAD, and provides support for the use of this approach in children with LAD who possess a histocompatible sibling donor.


Subject(s)
Disease Models, Animal , Hematopoietic Stem Cell Transplantation , Leukocyte-Adhesion Deficiency Syndrome/therapy , Animals , Dogs , Phenotype
12.
Pediatr Res ; 55(3): 363-7, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14711903

ABSTRACT

Children with the genetic immunodeficiency disease leukocyte adhesion deficiency, or LAD, develop life-threatening bacterial infections as a result of the inability of their leukocytes to adhere to the vessel wall and migrate to the sites of infection. Recently, the canine counterpart to LAD, known as canine leukocyte adhesion deficiency, or CLAD, has been described in Irish setter dogs. This review describes how the clinical phenotype of dogs with CLAD closely parallels that of children with the severe deficiency phenotype of LAD, thus enabling the CLAD dog to provide a disease-specific, large-animal model for testing novel hematopoietic stem cell and gene therapy strategies before their translation to children with LAD.


Subject(s)
Disease Models, Animal , Leukocyte-Adhesion Deficiency Syndrome/physiopathology , Animals , Child , Dogs , Genetic Therapy , Hematopoietic Stem Cell Transplantation , Humans , Leukocyte-Adhesion Deficiency Syndrome/therapy , Phenotype
13.
Blood ; 103(9): 3582-9, 2004 May 01.
Article in English | MEDLINE | ID: mdl-14715622

ABSTRACT

Children with the severe phenotype of the genetic immunodeficiency disease leukocyte adhesion deficiency or LAD experience life-threatening bacterial infections because of molecular defects in the leukocyte integrin CD18 molecule and the resultant failure to express the CD11/CD18 adhesion molecules on the leukocyte surface. Hematopoietic stem cell transplantation remains the only definitive therapy for LAD; however, the degree of donor chimerism and particularly the number of CD18(+) donor-derived neutrophils required to reverse the disease phenotype are not known. We performed nonmyeloablative hematopoietic stem cell transplantations from healthy matched littermates in 9 dogs with the canine form of LAD known as CLAD and demonstrate that in the 3 dogs with the lowest level of donor chimerism, less than 500 CD18(+) donor-derived neutrophils/microL in the peripheral blood of the CLAD recipients resulted in reversal of the CLAD disease phenotype. These results demonstrate the value of a disease-specific, large-animal model for identifying the lowest therapeutic level required for successful cellular and gene therapy.


Subject(s)
CD18 Antigens/analysis , Hematopoietic Stem Cell Transplantation , Leukocyte-Adhesion Deficiency Syndrome/therapy , Neutrophils/cytology , Animals , DNA/analysis , Dog Diseases , Dogs , Flow Cytometry , Leukocyte Count , Leukocyte-Adhesion Deficiency Syndrome/blood , Phenotype , Transplantation Chimera , Transplantation, Homologous , Treatment Outcome
14.
Vet Immunol Immunopathol ; 95(3-4): 113-21, 2003 Oct 15.
Article in English | MEDLINE | ID: mdl-12963272

ABSTRACT

The genetic disease canine leukocyte adhesion deficiency (CLAD) is characterized by recurrent, severe bacterial infections, typically culminating in death by 6 months of age. CLAD is due to a mutation in the leukocyte integrin CD18 subunit, which prevents surface expression of the CD11/CD18 leukocyte integrin complex. We demonstrate that stable mixed donor:host hematopoietic chimerism, achieved by a non-myeloablative bone marrow transplant from a histocompatible littermate, reverses the disease phenotype in CLAD. Donor chimerism following the transplant was demonstrated both by flow cytometric detection of donor-derived CD18-positive leukocytes in the peripheral blood of the recipient, and by the demonstration of donor-derived DNA microsatellite repeats in the peripheral blood leukocytes of the recipient. These results indicate that mixed hematopoietic chimerism reverses the clinical phenotype in CLAD and represents a potential therapeutic approach for the human disease leukocyte adhesion deficiency.


Subject(s)
Dog Diseases/therapy , Hematopoietic Stem Cell Transplantation/veterinary , Leukocyte-Adhesion Deficiency Syndrome/veterinary , Transplantation Chimera/immunology , Animals , Antigens, CD34/immunology , CD11 Antigens/immunology , CD18 Antigens/immunology , DNA/chemistry , DNA/genetics , Dog Diseases/immunology , Dog Diseases/pathology , Dogs , Flow Cytometry/veterinary , Hematopoietic Stem Cell Transplantation/methods , Leukocyte Count/veterinary , Leukocyte-Adhesion Deficiency Syndrome/immunology , Leukocyte-Adhesion Deficiency Syndrome/pathology , Leukocyte-Adhesion Deficiency Syndrome/therapy , Polymerase Chain Reaction/veterinary
15.
Vet Immunol Immunopathol ; 94(1-2): 11-22, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12842608

ABSTRACT

The genetic immunodeficiency disease canine leukocyte adhesion deficiency (CLAD) was originally described in juvenile Irish Setters with severe, recurrent bacterial infections. CLAD was subsequently shown to result from a mutation in the leukocyte integrin CD18 subunit which prevents leukocyte surface expression of the CD11/CD18 complex. We describe the development of a mixed-breed CLAD colony with clinical features that closely parallel those described in Irish Setters. We demonstrate that the early identification of CLAD heterozygotes and CLAD-affected dogs by a combination of flow cytometry and DNA sequencing allows the CLAD-affected animals to receive life-saving antibiotic therapy. The distinct clinical phenotype in CLAD, the ability to detect CD18 on the leukocyte surface by flow cytometry, and the history of the canine model in marrow transplantation, enable CLAD to serve as an attractive large-animal model for the investigation of novel hematopoietic stem cell and gene therapy strategies.


Subject(s)
Dog Diseases/genetics , Dogs/genetics , Leukocyte-Adhesion Deficiency Syndrome/genetics , Leukocyte-Adhesion Deficiency Syndrome/veterinary , Animals , Breeding , CD18 Antigens/analysis , Dog Diseases/pathology , Dog Diseases/therapy , Female , Genotype , Heterozygote , Leukocyte-Adhesion Deficiency Syndrome/pathology , Leukocyte-Adhesion Deficiency Syndrome/therapy , Male , Minisatellite Repeats/genetics , Mutation/genetics , Pedigree , Phenotype
16.
Blood ; 101(7): 2563-9, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12456496

ABSTRACT

The first human gene therapy experiment begun in September 1990 used a retroviral vector containing the human adenosine deaminase (ADA) cDNA to transduce mature peripheral blood lymphocytes from patients with ADA deficiency, an inherited disorder of immunity. Two patients who had been treated with intramuscular injections of pegylated bovine ADA (PEG-ADA) for 2 to 4 years were enrolled in this trial and each received a total of approximately 10(11) cells in 11 or 12 infusions over a period of about 2 years. No adverse events were observed. During and after treatment, the patients continued to receive PEG-ADA, although at a reduced dose. Ten years after the last cell infusion, approximately 20% of the first patient's lymphocytes still carry and express the retroviral gene, indicating that the effects of gene transfer can be remarkably long lasting. On the contrary, the persistence of gene-marked cells is very low (< 0.1%), and no expression of the transgene is detectable in lymphocytes from the second patient who developed persisting antibodies to components of the gene transfer system. Data collected from these original patients have provided novel information about the longevity of T lymphocytes in humans and persistence of gene expression in vivo from vectors driven by the Moloney murine leukemia virus long-terminal repeat (LTR) promoter. This long-term follow-up has also provided unique evidence supporting the safety of retroviral-mediated gene transfer and illustrates clear examples of both the potential and the pitfalls of gene therapy in humans.


Subject(s)
Adenosine Deaminase/deficiency , Adenosine Deaminase/genetics , Antibody Formation , Genetic Therapy/methods , Purine-Pyrimidine Metabolism, Inborn Errors/therapy , Adenosine Deaminase/administration & dosage , Adenosine Deaminase/biosynthesis , Animals , Antibodies, Heterophile/blood , Antibodies, Viral/blood , Cattle , Gene Expression , Gene Transfer Techniques , Genetic Vectors/immunology , Humans , Longitudinal Studies , Moloney murine leukemia virus/genetics , Moloney murine leukemia virus/immunology , Receptors, Antigen, T-Cell/analysis , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
17.
Hum Gene Ther ; 13(3): 425-32, 2002 Feb 10.
Article in English | MEDLINE | ID: mdl-11860709

ABSTRACT

Clinical gene therapy trials for adenosine deaminase (ADA) deficiency have shown limited success of corrective gene transfer into autologous T lymphocytes and CD34(+) cells. In these trials, the levels of gene transduction and expression in hematopoietic cells have been assessed by DNA- or RNA-based assays and measurement of ADA enzyme activity. Although informative, these methods are rarely applied to clonal analysis. The results of these assays therefore provide best estimates of transduction efficiency and gene expression in bulk populations based on the assumption that gene transfer and expression are uniformly distributed among transduced cells. As a useful additional tool for evaluation of ADA gene expression, we have developed a flow cytometry (fluorescence-activated cell sorting, FACS) assay capable of estimating the levels of intracellular ADA on a single-cell basis. We validated this technique with T cell lines and peripheral blood mononuclear cells (PBMCs) from ADA-deficient patients that showed severely reduced levels of ADA expression (ADA-dull) by FACS and Western blot analyses. After retrovirus-mediated ADA gene transfer, these cells showed clearly distinguishable populations exhibiting ADA expression (ADA-bright), thus allowing estimation of transduction efficiency. By mixing ADA-deficient and normal cells and using enzymatic amplification, we determined that our staining procedure could detect as little as 5% ADA-bright cells. This technique, therefore, will be useful to quickly assess the expression of ADA in hematopoietic cells of severe combined immunodeficient patients and represents an important tool for the follow-up of patients treated in clinical gene transfer protocols.


Subject(s)
Adenosine Deaminase/analysis , Adenosine Deaminase/genetics , Biological Assay , Genetic Therapy , Leukocytes, Mononuclear/enzymology , Severe Combined Immunodeficiency/diagnosis , Severe Combined Immunodeficiency/therapy , T-Lymphocytes/enzymology , Adenosine Deaminase/therapeutic use , Cell Line , Clinical Trials as Topic , Flow Cytometry/methods , Gene Expression Profiling , Human T-lymphotropic virus 1 , Humans , Sensitivity and Specificity , Severe Combined Immunodeficiency/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...