Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
EMBO J ; 42(23): e114188, 2023 Dec 01.
Article in English | MEDLINE | ID: mdl-37916874

ABSTRACT

Hyper IgM1 is an X-linked combined immunodeficiency caused by CD40LG mutations, potentially treatable with CD4+ T-cell gene editing with Cas9 and a "one-size-fits-most" corrective template. Contrary to established gene therapies, there is limited data on the genomic alterations following long-range gene editing, and no consensus on the relevant assays. We developed drop-off digital PCR assays for unbiased detection of large on-target deletions and found them at high frequency upon editing. Large deletions were also common upon editing different loci and cell types and using alternative Cas9 and template delivery methods. In CD40LG edited T cells, on-target deletions were counter-selected in culture and further purged by enrichment for edited cells using a selector coupled to gene correction. We then validated the sensitivity of optical genome mapping for unbiased detection of genome wide rearrangements and uncovered on-target trapping of one or more vector copies, which do not compromise functionality, upon editing using an integrase defective lentiviral donor template. No other recurring events were detected. Edited patient cells showed faithful reconstitution of CD40LG regulated expression and function with a satisfactory safety profile. Large deletions and donor template integrations should be anticipated and accounted for when designing and testing similar gene editing strategies.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Humans , Gene Editing/methods , Genome , T-Lymphocytes , CD4-Positive T-Lymphocytes
2.
Nat Commun ; 13(1): 2454, 2022 05 04.
Article in English | MEDLINE | ID: mdl-35508619

ABSTRACT

Liver gene therapy with adeno-associated viral (AAV) vectors delivering clotting factor transgenes into hepatocytes has shown multiyear therapeutic benefit in adults with hemophilia. However, the mostly episomal nature of AAV vectors challenges their application to young pediatric patients. We developed lentiviral vectors, which integrate in the host cell genome, that achieve efficient liver gene transfer in mice, dogs and non-human primates, by intravenous delivery. Here we first compare engineered coagulation factor VIII transgenes and show that codon-usage optimization improved expression 10-20-fold in hemophilia A mice and that inclusion of an unstructured XTEN peptide, known to increase the half-life of the payload protein, provided an additional >10-fold increase in overall factor VIII output in mice and non-human primates. Stable nearly life-long normal and above-normal factor VIII activity was achieved in hemophilia A mouse models. Overall, we show long-term factor VIII activity and restoration of hemostasis, by lentiviral gene therapy to hemophilia A mice and normal-range factor VIII activity in non-human primate, paving the way for potential clinical application.


Subject(s)
Hemophilia A , Animals , Child , Dogs , Factor VIII/genetics , Genetic Therapy , Genetic Vectors/genetics , Hemophilia A/genetics , Hemophilia A/therapy , Humans , Liver/metabolism , Mice , Primates/genetics
3.
Hum Gene Ther ; 32(1-2): 66-76, 2021 01.
Article in English | MEDLINE | ID: mdl-32883113

ABSTRACT

Hematopoietic stem cell gene therapy has become a successful therapeutic strategy for some inherited genetic disorders. Pre-clinical toxicity studies performed to support the human clinical trials using viral-mediated gene transfer and autologous hematopoietic stem and progenitor cell (HSPC) transplantation are complex and the use of mouse models of human diseases makes interpretation of the results challenging. In addition, they rely on the use of conditioning agents that must induce enough myeloablation to allow engraftment of transduced and transplanted HSPC. Busulfan and total body irradiation (TBI) are the most commonly used conditioning regimens in the mouse. Lenticular degeneration and atrophy of reproductive organs are expected histopathological changes. Proliferative and nonproliferative lesions can be observed with different incidence and distribution across strains and mouse models of diseases. The occurrence of these lesions can interfere with the interpretation of pre-clinical toxicity and tumorigenicity studies performed to support the human clinical studies. As such, it is important to be aware of the background incidence of lesions induced by different conditioning regimens. We review the histopathology results from seven long-term studies, five using TBI and two using busulfan.


Subject(s)
Hematopoietic Stem Cell Transplantation , Transplantation Conditioning , Animals , Busulfan , Cyclophosphamide , Genetic Therapy , Mice , Whole-Body Irradiation
4.
Mol Ther ; 29(1): 86-102, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33010230

ABSTRACT

Chronic granulomatous disease (CGD) is a rare inherited disorder due to loss-of-function mutations in genes encoding the NADPH oxidase subunits. Hematopoietic stem and progenitor cell (HSPC) gene therapy (GT) using regulated lentiviral vectors (LVs) has emerged as a promising therapeutic option for CGD patients. We performed non-clinical Good Laboratory Practice (GLP) and laboratory-grade studies to assess the safety and genotoxicity of LV targeting myeloid-specific Gp91phox expression in X-linked chronic granulomatous disease (XCGD) mice. We found persistence of gene-corrected cells for up to 1 year, restoration of Gp91phox expression and NADPH oxidase activity in XCGD phagocytes, and reduced tissue inflammation after LV-mediated HSPC GT. Although most of the mice showed no hematological or biochemical toxicity, a small subset of XCGD GT mice developed T cell lymphoblastic lymphoma (2.94%) and myeloid leukemia (5.88%). No hematological malignancies were identified in C57BL/6 mice transplanted with transduced XCGD HSPCs. Integration pattern analysis revealed an oligoclonal composition with rare dominant clones harboring vector insertions near oncogenes in mice with tumors. Collectively, our data support the long-term efficacy of LV-mediated HSPC GT in XCGD mice and provide a safety warning because the chronic inflammatory XCGD background may contribute to oncogenesis.


Subject(s)
Genetic Therapy , Genetic Vectors/genetics , Granulomatous Disease, Chronic/complications , Granulomatous Disease, Chronic/therapy , Hematologic Neoplasms/etiology , Lentivirus/genetics , Animals , Disease Models, Animal , Genetic Therapy/adverse effects , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Granulomatous Disease, Chronic/genetics , Humans , Mice , NADPH Oxidase 2/genetics , NADPH Oxidase 2/metabolism , Time Factors , Treatment Outcome
5.
Sci Transl Med ; 11(493)2019 05 22.
Article in English | MEDLINE | ID: mdl-31118293

ABSTRACT

Liver-directed gene therapy for the coagulation disorder hemophilia showed safe and effective results in clinical trials using adeno-associated viral vectors to replace a functional coagulation factor, although some unmet needs remain. Lentiviral vectors (LVs) may address some of these hurdles because of their potential for stable expression and the low prevalence of preexisting viral immunity in humans. However, systemic LV administration to hemophilic dogs was associated to mild acute toxicity and low efficacy at the administered doses. Here, exploiting intravital microscopy and LV surface engineering, we report a major role of the human phagocytosis inhibitor CD47, incorporated into LV cell membrane, in protecting LVs from uptake by professional phagocytes and innate immune sensing, thus favoring biodistribution to hepatocytes after systemic administration. By enforcing high CD47 surface content, we generated phagocytosis-shielded LVs which, upon intravenous administration to nonhuman primates, showed selective liver and spleen targeting and enhanced hepatocyte gene transfer compared to parental LV, reaching supraphysiological activity of human coagulation factor IX, the protein encoded by the transgene, without signs of toxicity or clonal expansion of transduced cells.


Subject(s)
Genetic Therapy , Genetic Vectors/therapeutic use , Lentivirus/genetics , Liver/pathology , Phagocytosis , Animals , CD47 Antigen/metabolism , Gene Transfer Techniques , Hepatocytes/metabolism , Humans , Immune Tolerance , Immunity, Innate , Kupffer Cells/metabolism , Macaca , Mice, Inbred C57BL , Mice, Inbred NOD , Phagocytes/metabolism , Tissue Distribution
6.
Hum Gene Ther ; 27(10): 813-829, 2016 10.
Article in English | MEDLINE | ID: mdl-27431943

ABSTRACT

In order to support the clinical application of hematopoietic stem cell (HSC) gene therapy for mucopolysaccharidosis I (MPS I), biosafety studies were conducted to assess the toxicity and tumorigenic potential, as well as the biodistribution of HSCs and progenitor cells (HSPCs) transduced with lentiviral vectors (LV) encoding the cDNA of the alpha-iduronidase (IDUA) gene, which is mutated in MPS I patients. To this goal, toxicology and biodistribution studies were conducted, employing Good Laboratory Practice principles. Vector integration site (IS) studies were applied in order to predict adverse consequences of vector gene transfer and to obtain HSC-related information. Overall, the results obtained in these studies provided robust evidence to support the safety and tolerability of high-efficiency LV-mediated gene transfer and above-normal IDUA enzyme expression in both murine and human HSPCs and their in vivo progeny. Taken together, these investigations provide essential safety data to support clinical testing of HSC gene therapy in MPS I patients. These studies also underline criticisms associated with the use of currently available models, and highlight the value of surrogate markers of tumorigenicity that may be further explored in the future. Notably, biological evidence supporting the efficacy of gene therapy on MPS I disease and its feasibility on patients' HSCs were also generated, employing clinical-grade LVs. Finally, the clonal contribution of LV-transduced HSPCs to hematopoiesis along serial transplantation was quantified in a minimum of 200-300 clones, with the different level of repopulating cells in primary recipients being reflected in the secondary.


Subject(s)
Genetic Therapy/adverse effects , Hematopoietic Stem Cell Transplantation/adverse effects , Iduronidase/genetics , Mucopolysaccharidosis I/therapy , Animals , Gene Expression Regulation, Enzymologic , Gene Transfer Techniques/adverse effects , Genetic Therapy/methods , Genetic Vectors/adverse effects , Hematopoietic Stem Cells/enzymology , Humans , Iduronidase/adverse effects , Iduronidase/therapeutic use , Lentivirus/genetics , Mice , Mucopolysaccharidosis I/genetics
7.
Curr Opin Hematol ; 18(4): 214-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21537167

ABSTRACT

PURPOSE OF REVIEW: The maintenance of a functional hematopoietic niche is critical for modulating the fate of hematopoietic stem cells (HSCs). Several enzymes were described as essential for guaranteeing niche functionality. This review summarizes the recent findings about the role of galactocerebrosidase and other enzymes involved in the maintenance of a functional HSC niche. RECENT FINDINGS: The essential role of enzymes actively involved in the maintenance of the bone marrow microenvironment, in bone remodeling, in regulating the sympathetic innervation of the niche, and in the production and relative balance of sphingolipids active in the niche has been recently highlighted. Enzymes involved in bone remodeling modify the cell-to-cell interaction between osteoblasts and HSCs. Heparanase, neutrophil elastase, and alpha-iduronidase affect the bioavailability of key cytokines and ligands within the extracellular matrix of the niche. Moreover, galactosyltransferase and galactocerebrosidase affect the function of the sympathetic nervous system and/or the balance of bioactive sphingolipids, thus influencing the SDF-1/CXCR4 axis and the proliferation of HSCs. SUMMARY: Here, we discuss the role of different enzymes directly or indirectly influencing the niche microenvironment, and we provide a comprehensive picture of their cooperative role, together with receptors, soluble factors, and the extracellular matrix, in maintaining a functional hematopoietic niche.


Subject(s)
Galactosylceramidase/metabolism , Hematopoietic Stem Cells/enzymology , Stem Cell Niche/enzymology , Animals , Hematopoietic Stem Cells/cytology , Humans , Mice , Stem Cell Niche/cytology
8.
Sci Transl Med ; 2(58): 58ra84, 2010 Nov 17.
Article in English | MEDLINE | ID: mdl-21084719

ABSTRACT

Globoid cell leukodystrophy (GLD; also known as Krabbe disease) is an invariably fatal lysosomal storage disorder caused by mutations in the galactocerebrosidase (GALC) gene. Hematopoietic stem cell (HSC)-based gene therapy is being explored for GLD; however, we found that forced GALC expression was toxic to HSCs and early progenitors, highlighting the need for improved regulation of vector expression. We used a genetic reporter strategy based on lentiviral vectors to detect microRNA activity in hematopoietic cells at single-cell resolution. We report that miR-126 and miR-130a were expressed in HSCs and early progenitors from both mice and humans, but not in differentiated progeny. Moreover, repopulating HSCs could be purified solely on the basis of miRNA expression, providing a new method relevant for human HSC isolation. By incorporating miR-126 target sequences into a GALC-expressing vector, we suppressed GALC expression in HSCs while maintaining robust expression in mature hematopoietic cells. This approach protected HSCs from GALC toxicity and allowed successful treatment of a mouse GLD model, providing a rationale to explore HSC-based gene therapy for GLD.


Subject(s)
Genetic Therapy , Hematopoietic Stem Cells/metabolism , Leukodystrophy, Globoid Cell/genetics , Leukodystrophy, Globoid Cell/therapy , MicroRNAs/metabolism , Animals , Cell Differentiation , Cell Separation , Cytoprotection , Galactosylceramidase/metabolism , Gene Expression Regulation , Genes, Transgenic, Suicide , Hematopoietic Stem Cells/cytology , Humans , Mice , Mice, SCID , MicroRNAs/genetics , Organ Specificity/genetics
9.
Blood ; 116(24): 5130-9, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-20847202

ABSTRACT

Type I mucopolysaccharidosis (MPS I) is a lysosomal storage disorder caused by the deficiency of α-L-iduronidase, which results in glycosaminoglycan accumulation in tissues. Clinical manifestations include skeletal dysplasia, joint stiffness, visual and auditory defects, cardiac insufficiency, hepatosplenomegaly, and mental retardation (the last being present exclusively in the severe Hurler variant). The available treatments, enzyme-replacement therapy and hematopoietic stem cell (HSC) transplantation, can ameliorate most disease manifestations, but their outcome on skeletal and brain disease could be further improved. We demonstrate here that HSC gene therapy, based on lentiviral vectors, completely corrects disease manifestations in the mouse model. Of note, the therapeutic benefit provided by gene therapy on critical MPS I manifestations, such as neurologic and skeletal disease, greatly exceeds that exerted by HSC transplantation, the standard of care treatment for Hurler patients. Interestingly, therapeutic efficacy of HSC gene therapy is strictly dependent on the achievement of supranormal enzyme activity in the hematopoietic system of transplanted mice, which allows enzyme delivery to the brain and skeleton for disease correction. Overall, our data provide evidence of an efficacious treatment for MPS I Hurler patients, warranting future development toward clinical testing.


Subject(s)
Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation/methods , Iduronidase/administration & dosage , Mucopolysaccharidosis I/therapy , Animals , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology , Brain/drug effects , Brain/metabolism , Brain/pathology , Disease Models, Animal , Genetic Vectors , Iduronidase/genetics , Lentivirus/genetics , Mice , Mice, Knockout , Mucopolysaccharidosis I/pathology , Phenotype , Remission Induction , Treatment Outcome
10.
J Exp Med ; 207(8): 1647-60, 2010 Aug 02.
Article in English | MEDLINE | ID: mdl-20643830

ABSTRACT

Self-renewal and differentiation of hematopoietic stem cells (HSCs) are balanced by the concerted activities of the fibroblast growth factor (FGF), Wnt, and Notch pathways, which are tuned by enzyme-mediated remodeling of heparan sulfate proteoglycans (HSPGs). Sulfatase modifying factor 1 (SUMF1) activates the Sulf1 and Sulf2 sulfatases that remodel the HSPGs, and is mutated in patients with multiple sulfatase deficiency. Here, we show that the FGF signaling pathway is constitutively activated in Sumf1(-/-) HSCs and hematopoietic stem progenitor cells (HSPCs). These cells show increased p-extracellular signal-regulated kinase levels, which in turn promote beta-catenin accumulation. Constitutive activation of FGF signaling results in a block in erythroid differentiation at the chromatophilic erythroblast stage, and of B lymphocyte differentiation at the pro-B cell stage. A reduction in mature myeloid cells and an aberrant development of T lymphocytes are also seen. These defects are rescued in vivo by blocking the FGF pathway in Sumf1(-/-) mice. Transplantation of Sumf1(-/-) HSPCs into wild-type mice reconstituted the phenotype of the donors, suggesting a cell autonomous defect. These data indicate that Sumf1 controls HSPC differentiation and hematopoietic lineage development through FGF and Wnt signaling.


Subject(s)
Fibroblast Growth Factors/physiology , Hematopoiesis/physiology , Signal Transduction/physiology , Sulfatases/genetics , Animal Structures/cytology , Animals , Antigens, CD/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Butadienes/pharmacology , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation/drug effects , Cell Differentiation/physiology , Colony-Forming Units Assay , Erythropoiesis/drug effects , Erythropoiesis/physiology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factors/antagonists & inhibitors , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Gene Expression/drug effects , Gene Expression/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hematopoiesis/drug effects , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Lymphopoiesis/drug effects , Lymphopoiesis/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelopoiesis/drug effects , Myelopoiesis/physiology , Nitriles/pharmacology , Oxidoreductases Acting on Sulfur Group Donors , Phosphorylation/drug effects , Phosphorylation/genetics , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Notch1/genetics , Signal Transduction/drug effects , Stem Cells/cytology , Stem Cells/metabolism , Sulfatases/metabolism , Sulfotransferases/genetics , Sulfotransferases/metabolism , Wnt Proteins/physiology , beta Catenin/metabolism
11.
Blood ; 116(11): 1857-66, 2010 Sep 16.
Article in English | MEDLINE | ID: mdl-20511539

ABSTRACT

The balance between survival and death in many cell types is regulated by small changes in the intracellular content of bioactive sphingolipids. Enzymes that either produce or degrade these sphingolipids control this equilibrium. The findings here described indicate that the lysosomal galactocerebrosidase (GALC) enzyme, defective in globoid cell leukodystrophy, is involved in the maintenance of a functional hematopoietic stem/progenitor cell (HSPC) niche by contributing to the control of the intracellular content of key sphingolipids. Indeed, we show that both insufficient and supraphysiologic GALC activity-by inherited genetic deficiency or forced gene expression in patients' cells and in the disease model-induce alterations of the intracellular content of the bioactive GALC downstream products ceramide and sphingosine, and thus affect HSPC survival and function and the functionality of the stem cell niche. Therefore, GALC and, possibly, other enzymes for the maintenance of niche functionality and health tightly control the concentration of these sphingolipids within HSPCs.


Subject(s)
Bone Marrow/enzymology , Galactosylceramidase/metabolism , Hematopoietic Stem Cells/enzymology , Stem Cell Niche/enzymology , Animals , Apoptosis/drug effects , Bone Marrow/metabolism , Cell Survival/drug effects , Cells, Cultured , Flow Cytometry , Galactosylceramidase/deficiency , Galactosylceramidase/genetics , Genotype , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Humans , Immunophenotyping , In Situ Nick-End Labeling , Insulin-Like Growth Factor I/pharmacology , Leukodystrophy, Globoid Cell/enzymology , Leukodystrophy, Globoid Cell/genetics , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Sphingolipids/metabolism , Stem Cell Niche/metabolism , Transfection , U937 Cells
12.
Eur J Immunol ; 39(10): 2748-54, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19637231

ABSTRACT

A defect in invariant NKT (iNKT) cell selection was hypothesized in lysosomal storage disorders (LSD). Accumulation of glycosphingolipids (GSL) in LSD could influence lipid loading and/or presentation causing entrapment of endogenous ligand(s) within storage bodies or competition of the selecting ligand(s) by stored lipids for CD1d binding. However, when we analyzed the iNKT cell compartment in newly tested LSD animal models that accumulate GSL, glycoaminoglycans or both, we observed a defective iNKT cell selection only in animals affected by multiple sulfatase deficiency, in which a generalized aberrant T-cell development, rather than a pure iNKT defect, was present. Mice with single lysosomal enzyme deficiencies had normal iNKT cell development. Thus, GSL/glycoaminoglycans storage and lysosomal engulfment are not sufficient for affecting iNKT cell development. Rather, lipid ligand(s) or storage compounds, which are affected in those LSD lacking mature iNKT cells, might indeed be relevant for iNKT cell selection.


Subject(s)
Cell Differentiation/immunology , Lysosomal Storage Diseases/immunology , Natural Killer T-Cells/immunology , Animals , Cell Count , Cell Differentiation/drug effects , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Female , Leukodystrophy, Globoid Cell/genetics , Leukodystrophy, Globoid Cell/immunology , Leukodystrophy, Globoid Cell/pathology , Leukodystrophy, Metachromatic/genetics , Leukodystrophy, Metachromatic/immunology , Liver/immunology , Liver/pathology , Lymphocytes/pathology , Lysosomal Storage Diseases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mucopolysaccharidosis I/genetics , Mucopolysaccharidosis I/immunology , Multiple Sulfatase Deficiency Disease/genetics , Multiple Sulfatase Deficiency Disease/immunology , Multiple Sulfatase Deficiency Disease/pathology , Natural Killer T-Cells/pathology , Pyrroles/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Sandhoff Disease/genetics , Sandhoff Disease/immunology , Sandhoff Disease/pathology , Spleen/immunology , Spleen/pathology , Thymus Gland/immunology , Thymus Gland/pathology
13.
J Neurosci ; 29(27): 8858-70, 2009 Jul 08.
Article in English | MEDLINE | ID: mdl-19587293

ABSTRACT

How membrane biosynthesis and homeostasis is achieved in myelinating glia is mostly unknown. We previously reported that loss of myotubularin-related protein 2 (MTMR2) provokes autosomal recessive demyelinating Charcot-Marie-Tooth type 4B1 neuropathy, characterized by excessive redundant myelin, also known as myelin outfoldings. We generated a Mtmr2-null mouse that models the human neuropathy. We also found that, in Schwann cells, Mtmr2 interacts with Discs large 1 (Dlg1), a scaffold involved in polarized trafficking and membrane addition, whose localization in Mtmr2-null nerves is altered. We here report that, in Schwann cells, Dlg1 also interacts with kinesin 13B (kif13B) and Sec8, which are involved in vesicle transport and membrane tethering in polarized cells, respectively. Taking advantage of the Mtmr2-null mouse as a model of impaired membrane formation, we provide here the first evidence for a machinery that titrates membrane formation during myelination. We established Schwann cell/DRG neuron cocultures from Mtmr2-null mice, in which myelin outfoldings were reproduced and almost completely rescued by Mtmr2 replacement. By exploiting this in vitro model, we propose a mechanism whereby kif13B kinesin transports Dlg1 to sites of membrane remodeling where it coordinates a homeostatic control of myelination. The interaction of Dlg1 with the Sec8 exocyst component promotes membrane addition, whereas with Mtmr2, negatively regulates membrane formation. Myelin outfoldings thus arise as a consequence of the loss of negative control on the amount of membrane, which is produced during myelination.


Subject(s)
Carrier Proteins/physiology , Cell Membrane/physiology , Homeostasis/physiology , Myelin Sheath/physiology , Nerve Tissue Proteins/physiology , Protein Tyrosine Phosphatases, Non-Receptor/physiology , Schwann Cells/physiology , Animals , COS Cells , Carrier Proteins/metabolism , Cells, Cultured , Chlorocebus aethiops , Coculture Techniques , Discs Large Homolog 1 Protein , Down-Regulation/physiology , Exocytosis/physiology , HeLa Cells , Humans , Membrane Proteins , Mice , Mice, Knockout , Nerve Fibers, Myelinated/physiology , Nerve Tissue Proteins/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/deficiency , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Rats , SAP90-PSD95 Associated Proteins , Schwann Cells/cytology
14.
Neurobiol Dis ; 34(1): 51-62, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19320046

ABSTRACT

Microglia activation and neuroinflammation play a pivotal role in the pathogenesis of lysosomal storage disorders (LSD) affecting the central nervous system (CNS), which are amenable to treatment by hematopoietic stem cell transplantation (HSCT). HSCT efficacy relies on replacing the intra- and extra-vascular hematopoietic cell compartments, including CNS microglia, with a cell population expressing the functional enzyme. Non-invasive and quantitative assessment of microglia activation and of its reduction upon HSCT might allow for evaluation of disease evolution and response to treatment in LSD. We here demonstrate that microglia activation can be quantified ex vivo and in vivo by PET using the peripheral benzodiazepine receptor ligand PK11195 in two models of LSD. Furthermore, we show a differential PBR binding following microglia replacement by donor cells in mice undergoing HSCT. Our data indicates that PBR ligands constitute valuable tools for monitoring the evolution and the response to treatment of LSD with CNS involvement, and enable us to evaluate whether the turnover between endogenous and donor microglia following HSCT could be adequate enough to delay disease progression.


Subject(s)
Central Nervous System Diseases/physiopathology , Hematopoietic Stem Cell Transplantation , Isoquinolines/metabolism , Lysosomal Storage Diseases, Nervous System/physiopathology , Microglia/physiology , Receptors, GABA-A/metabolism , Animals , Brain/diagnostic imaging , Brain/physiopathology , Carbon Radioisotopes , Central Nervous System Diseases/diagnostic imaging , Central Nervous System Diseases/therapy , Demyelinating Diseases/physiopathology , Disease Models, Animal , Fluorescent Antibody Technique , Gliosis/physiopathology , Ligands , Lysosomal Storage Diseases, Nervous System/diagnostic imaging , Lysosomal Storage Diseases, Nervous System/therapy , Mice , Mice, Transgenic , Microscopy, Confocal , Positron-Emission Tomography
15.
Clin Chem ; 55(3): 541-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19147730

ABSTRACT

BACKGROUND: The determination of cellular beta-galactocerebrosidase activity is an established procedure to diagnose Krabbe disease and monitor the efficacy of gene/stem cell-based therapeutic approaches aimed at restoring defective enzymatic activity in patients or disease models. Current biochemical assays for beta-galactocerebrosidase show high specificity but generally require large protein amounts from scanty sources such as hematopoietic or neural stem cells. We developed a novel assay based on the hypothesis that specific measurements of beta-galactocerebrosidase activity can be performed following complete inhibition of beta-galactosidase activity. METHODS: We performed the assay using 2-7.5 microg of sample proteins with the artificial fluorogenic substrate 4-methylumbelliferone-beta-galactopyranoside (1.5 mmol/L) resuspended in 0.1/0.2 mol/L citrate/phosphate buffer, pH 4.0, and AgNO(3). Reactions were incubated for 30 min at 37 degrees C. Fluorescence of liberated 4-methylumbelliferone was measured on a spectrofluorometer (lambda(ex) 360 nm, lambda(em) 446 nm). RESULTS: AgNO(3) was a competitive inhibitor of beta-galactosidase [inhibition constant (K(i)) = 0.12 micromol/L] and completely inhibited beta-galactosidase activity when used at a concentration of 11 micromol/L. Under this condition, the beta-galactocerebrosidase activity was preserved and could be specifically and accurately measured. The assay can detect beta-galactocerebrosidase activity in as little as 2 microg cell protein extract or 7.5 microg tissue. Assay validation was performed using (a) brain tissues from wild-type and twitcher mice and (b) murine GALC(-/-) hematopoietic stem cells and neural precursor cells transduced by GALC-lentiviral vectors. CONCLUSIONS: The procedure is straightforward, rapid, and reproducible. Within a clinical context, our method unequivocally discriminated cells from healthy subjects and Krabbe patients and is therefore suitable for diagnostic applications.


Subject(s)
Galactosylceramidase/analysis , Galactosylceramidase/metabolism , beta-Galactosidase/antagonists & inhibitors , beta-Galactosidase/metabolism , Animals , Cells, Cultured , Chromatography, Gel , Enzyme Activation/drug effects , Humans , Mice , Mice, Inbred C57BL , Silver Nitrate/pharmacology , Tissue Culture Techniques
16.
Biochem J ; 403(2): 305-12, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17206939

ABSTRACT

Sulfatases are enzymes that hydrolyse a diverse range of sulfate esters. Deficiency of lysosomal sulfatases leads to human diseases characterized by the accumulation of either GAGs (glycosaminoglycans) or sulfolipids. The catalytic activity of sulfatases resides in a unique formylglycine residue in their active site generated by the post-translational modification of a highly conserved cysteine residue. This modification is performed by SUMF1 (sulfatase-modifying factor 1), which is an essential factor for sulfatase activities. Mutations in the SUMF1 gene cause MSD (multiple sulfatase deficiency), an autosomal recessive disease in which the activities of all sulfatases are profoundly reduced. In previous studies, we have shown that SUMF1 has an enhancing effect on sulfatase activity when co-expressed with sulfatase genes in COS-7 cells. In the present study, we demonstrate that SUMF1 displays an enhancing effect on sulfatases activity when co-delivered with a sulfatase cDNA via AAV (adeno-associated virus) and LV (lentivirus) vectors in cells from individuals affected by five different diseases owing to sulfatase deficiencies or from murine models of the same diseases [i.e. MLD (metachromatic leukodystrophy), CDPX (X-linked dominant chondrodysplasia punctata) and MPS (mucopolysaccharidosis) II, IIIA and VI]. The SUMF1-enhancing effect on sulfatase activity resulted in an improved clearance of the intracellular GAG or sulfolipid accumulation. Moreover, we demonstrate that the SUMF1-enhancing effect is also present in vivo after AAV-mediated delivery of the sulfamidase gene to the muscle of MPSIIIA mice, resulting in a more efficient rescue of the phenotype. These results indicate that co-delivery of SUMF1 may enhance the efficacy of gene therapy in several sulfatase deficiencies.


Subject(s)
Sulfatases/deficiency , Sulfatases/metabolism , Adenoviridae/genetics , Animals , Cells, Cultured , Cysteine/genetics , Cysteine/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Lentivirus/genetics , Lentivirus/metabolism , Male , Mice , Muscles/enzymology , Mutation/genetics , Oxidoreductases Acting on Sulfur Group Donors , Protein Transport , Sulfatases/genetics
17.
J Clin Invest ; 113(8): 1118-29, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15085191

ABSTRACT

Gene-based delivery can establish a sustained supply of therapeutic proteins within the nervous system. For diseases characterized by extensive CNS and peripheral nervous system (PNS) involvement, widespread distribution of the exogenous gene may be required, a challenge to in vivo gene transfer strategies. Here, using lentiviral vectors (LVs), we efficiently transduced hematopoietic stem cells (HSCs) ex vivo and evaluated the potential of their progeny to target therapeutic genes to the CNS and PNS of transplanted mice and correct a neurodegenerative disorder, metachromatic leukodystrophy (MLD). We proved extensive repopulation of CNS microglia and PNS endoneurial macrophages by transgene-expressing cells. Intriguingly, recruitment of these HSC-derived cells was faster and more robust in MLD mice. By transplanting HSCs transduced with the arylsulfatase A gene, we fully reconstituted enzyme activity in the hematopoietic system of MLD mice and prevented the development of motor conduction impairment, learning and coordination deficits, and neuropathological abnormalities typical of the disease. Remarkably, ex vivo gene therapy had a significantly higher therapeutic impact than WT HSC transplantation, indicating a critical role for enzyme overexpression in the HSC progeny. These results indicate that transplantation of LV-transduced autologous HSCs represents a potentially efficacious therapeutic strategy for MLD and possibly other neurodegenerative disorders.


Subject(s)
Genetic Therapy , Hematopoietic Stem Cell Transplantation , Leukodystrophy, Metachromatic/therapy , Animals , Cell Differentiation , Cell Movement , Disease Models, Animal , Lentivirus/genetics , Mice , Mice, Inbred C57BL , Motor Activity , Nervous System/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...