Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
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
2.
Nat Commun ; 9(1): 2896, 2018 07 24.
Article in English | MEDLINE | ID: mdl-30042420

ABSTRACT

Immunotherapy is emerging as a new pillar of cancer treatment with potential to cure. However, many patients still fail to respond to these therapies. Among the underlying factors, an immunosuppressive tumor microenvironment (TME) plays a major role. Here we show that monocyte-mediated gene delivery of IFNα inhibits leukemia in a mouse model. IFN gene therapy counteracts leukemia-induced expansion of immunosuppressive myeloid cells and imposes an immunostimulatory program to the TME, as shown by bulk and single-cell transcriptome analyses. This reprogramming promotes T-cell priming and effector function against multiple surrogate tumor-specific antigens, inhibiting leukemia growth in our experimental model. Durable responses are observed in a fraction of mice and are further increased combining gene therapy with checkpoint blockers. Furthermore, IFN gene therapy strongly enhances anti-tumor activity of adoptively transferred T cells engineered with tumor-specific TCR or CAR, overcoming suppressive signals in the leukemia TME. These findings warrant further investigations on the potential development of our gene therapy strategy towards clinical testing.


Subject(s)
Antigens, Neoplasm/immunology , Genetic Therapy/methods , Immunity/immunology , Interferons/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Tumor Microenvironment/immunology , Animals , Cell Line, Tumor , Cells, Cultured , Female , Gene Expression Regulation, Leukemic , Immunity/genetics , Immunotherapy, Adoptive/methods , Interferons/genetics , Interferons/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , Tumor Microenvironment/genetics
3.
Sci Transl Med ; 9(411)2017 Oct 11.
Article in English | MEDLINE | ID: mdl-29021165

ABSTRACT

Targeted genome editing in hematopoietic stem/progenitor cells (HSPCs) is an attractive strategy for treating immunohematological diseases. However, the limited efficiency of homology-directed editing in primitive HSPCs constrains the yield of corrected cells and might affect the feasibility and safety of clinical translation. These concerns need to be addressed in stringent preclinical models and overcome by developing more efficient editing methods. We generated a humanized X-linked severe combined immunodeficiency (SCID-X1) mouse model and evaluated the efficacy and safety of hematopoietic reconstitution from limited input of functional HSPCs, establishing thresholds for full correction upon different types of conditioning. Unexpectedly, conditioning before HSPC infusion was required to protect the mice from lymphoma developing when transplanting small numbers of progenitors. We then designed a one-size-fits-all IL2RG (interleukin-2 receptor common γ-chain) gene correction strategy and, using the same reagents suitable for correction of human HSPC, validated the edited human gene in the disease model in vivo, providing evidence of targeted gene editing in mouse HSPCs and demonstrating the functionality of the IL2RG-edited lymphoid progeny. Finally, we optimized editing reagents and protocol for human HSPCs and attained the threshold of IL2RG editing in long-term repopulating cells predicted to safely rescue the disease, using clinically relevant HSPC sources and highly specific zinc finger nucleases or CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 (CRISPR-associated protein 9). Overall, our work establishes the rationale and guiding principles for clinical translation of SCID-X1 gene editing and provides a framework for developing gene correction for other diseases.


Subject(s)
Hematopoietic Stem Cells/metabolism , Animals , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gene Editing/methods , Gene Targeting/methods , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/metabolism , Mice , Mice, SCID
4.
Cancer Cell ; 29(6): 905-921, 2016 06 13.
Article in English | MEDLINE | ID: mdl-27300437

ABSTRACT

MicroRNA (miRNA)-126 is a known regulator of hematopoietic stem cell quiescence. We engineered murine hematopoiesis to express miRNA-126 across all differentiation stages. Thirty percent of mice developed monoclonal B cell leukemia, which was prevented or regressed when a tetracycline-repressible miRNA-126 cassette was switched off. Regression was accompanied by upregulation of cell-cycle regulators and B cell differentiation genes, and downregulation of oncogenic signaling pathways. Expression of dominant-negative p53 delayed blast clearance upon miRNA-126 switch-off, highlighting the relevance of p53 inhibition in miRNA-126 addiction. Forced miRNA-126 expression in mouse and human progenitors reduced p53 transcriptional activity through regulation of multiple p53-related targets. miRNA-126 is highly expressed in a subset of human B-ALL, and antagonizing miRNA-126 in ALL xenograft models triggered apoptosis and reduced disease burden.


Subject(s)
Hematopoietic Stem Cells/metabolism , MicroRNAs/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Tumor Suppressor Protein p53/genetics , Animals , Apoptosis , Cell Cycle , Cell Differentiation , Gene Expression Regulation, Neoplastic , Hematopoietic Stem Cell Transplantation , Humans , Mice , MicroRNAs/metabolism , Neoplasms, Experimental , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Signal Transduction , Up-Regulation
5.
Exp Hematol ; 43(10): 858-868.e7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26163797

ABSTRACT

A precise understanding of the role of miR-223 in human hematopoiesis and in the pathogenesis of acute myeloid leukemia (AML) is still lacking. By measuring miR-223 expression in blasts from 115 AML patients, we found significantly higher miR-223 levels in patients with favorable prognosis, whereas patients with low miR-223 expression levels were associated with worse outcome. Furthermore, miR-223 was hierarchically expressed in AML subpopulations, with lower expression in leukemic stem cell-containing fractions. Genetic depletion of miR-223 decreased the leukemia initiating cell (LIC) frequency in a myelomonocytic AML mouse model, but it was not mandatory for rapid-onset AML. To relate these observations to physiologic myeloid differentiation, we knocked down or ectopically expressed miR-223 in cord-blood CD34⁺ cells using lentiviral vectors. Although miR-223 knockdown delayed myeloerythroid precursor differentiation in vitro, it increased myeloid progenitors in vivo following serial xenotransplantation. Ectopic miR-223 expression increased erythropoiesis, T lymphopoiesis, and early B lymphopoiesis in vivo. These findings broaden the role of miR-223 as a regulator of the expansion/differentiation equilibrium in hematopoietic stem and progenitor cells where its impact is dose- and differentiation-stage-dependent. This also explains the complex yet minor role of miR-223 in AML, a heterogeneous disease with variable degree of myeloid differentiation.


Subject(s)
Fetal Blood/metabolism , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/metabolism , MicroRNAs/biosynthesis , Neoplasms, Experimental/metabolism , Neoplastic Stem Cells/metabolism , RNA, Neoplasm/biosynthesis , Adult , Animals , Cell Proliferation/genetics , Erythropoiesis/genetics , Female , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Lymphopoiesis/genetics , Male , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , MicroRNAs/genetics , Middle Aged , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Neoplastic Stem Cells/pathology , RNA, Neoplasm/genetics
6.
Science ; 341(6148): 1233158, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23845948

ABSTRACT

Metachromatic leukodystrophy (MLD) is an inherited lysosomal storage disease caused by arylsulfatase A (ARSA) deficiency. Patients with MLD exhibit progressive motor and cognitive impairment and die within a few years of symptom onset. We used a lentiviral vector to transfer a functional ARSA gene into hematopoietic stem cells (HSCs) from three presymptomatic patients who showed genetic, biochemical, and neurophysiological evidence of late infantile MLD. After reinfusion of the gene-corrected HSCs, the patients showed extensive and stable ARSA gene replacement, which led to high enzyme expression throughout hematopoietic lineages and in cerebrospinal fluid. Analyses of vector integrations revealed no evidence of aberrant clonal behavior. The disease did not manifest or progress in the three patients 7 to 21 months beyond the predicted age of symptom onset. These findings indicate that extensive genetic engineering of human hematopoiesis can be achieved with lentiviral vectors and that this approach may offer therapeutic benefit for MLD patients.


Subject(s)
Cerebroside-Sulfatase/genetics , Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Leukodystrophy, Metachromatic/therapy , Brain/pathology , DNA Damage , Follow-Up Studies , Genetic Engineering , Genetic Vectors/toxicity , Humans , Lentivirus , Leukodystrophy, Metachromatic/pathology , Magnetic Resonance Imaging , Transduction, Genetic , Treatment Outcome , Virus Integration
7.
Proc Natl Acad Sci U S A ; 109(37): 15018-23, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22923692

ABSTRACT

The recent hypothesis that postnatal microglia are maintained independently of circulating monocytes by local precursors that colonize the brain before birth has relevant implications for the treatment of various neurological diseases, including lysosomal storage disorders (LSDs), for which hematopoietic cell transplantation (HCT) is applied to repopulate the recipient myeloid compartment, including microglia, with cells expressing the defective functional hydrolase. By studying wild-type and LSD mice at diverse time-points after HCT, we showed the occurrence of a short-term wave of brain infiltration by a fraction of the transplanted hematopoietic progenitors, independently from the administration of a preparatory regimen and from the presence of a disease state in the brain. However, only the use of a conditioning regimen capable of ablating functionally defined brain-resident myeloid precursors allowed turnover of microglia with the donor, mediated by local proliferation of early immigrants rather than entrance of mature cells from the circulation.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Lysosomal Storage Diseases, Nervous System/therapy , Microglia/cytology , Transplantation Conditioning/methods , Analysis of Variance , Animals , Cell Differentiation/physiology , Cell Movement/physiology , Flow Cytometry , Green Fluorescent Proteins/metabolism , In Situ Nick-End Labeling , Mice , Mice, Knockout
8.
Blood ; 117(20): 5332-9, 2011 May 19.
Article in English | MEDLINE | ID: mdl-21403130

ABSTRACT

A recent clinical trial for adrenoleukodystrophy (ALD) showed the efficacy and safety of lentiviral vector (LV) gene transfer in hematopoietic stem progenitor cells. However, several common insertion sites (CIS) were found in patients' cells, suggesting that LV integrations conferred a selective advantage. We performed high-throughput LV integration site analysis on human hematopoietic stem progenitor cells engrafted in immunodeficient mice and found the same CISs reported in patients with ALD. Strikingly, most CISs in our experimental model and in patients with ALD cluster in megabase-wide chromosomal regions of high LV integration density. Conversely, cancer-triggering integrations at CISs found in tumor cells from γ-retroviral vector-based clinical trials and oncogene-tagging screenings in mice always target a single gene and are contained in narrow genomic intervals. These findings imply that LV CISs are produced by an integration bias toward specific genomic regions rather than by oncogenic selection.


Subject(s)
Genetic Therapy/methods , Genetic Vectors , Lentivirus/genetics , Adrenoleukodystrophy/genetics , Adrenoleukodystrophy/therapy , Animals , Clinical Trials as Topic , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Gene Transfer Techniques , Hematopoietic Stem Cell Transplantation , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Mice , Mice, Knockout , Transplantation Chimera/genetics , Virus Integration/genetics
9.
Hum Mutat ; 30(10): E936-45, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19606494

ABSTRACT

Metachromatic Leukodystrophy (MLD) is a rare inherited lysosomal storage disorder caused by the deficiency of Arylsulfatase A (ARSA). The disease manifests itself with a broad spectrum of clinical variants, all characterized by progressive neurodegeneration in the central and peripheral nervous systems. The correlation between mutations in the ARSA gene, residual enzymatic activity associated with the mutated alleles and patients' phenotype, which has been extensively drawn for common ARSA mutations, has recently been expanded to rare ones. In this context, functional studies on the rare allelic variances acquire particular relevance for patients' prognostic evaluation. Here we have characterized eight newly identified ARSA mutations, through lentiviral vector-based expression studies on cell lines and ARSA defective murine fibroblasts. In each case, the residual activity associated with the new mutant allele correlates well with the patient's phenotype. Therefore, our results confirm the importance of functional characterization of mutant alleles for a precise genotype-based classification and definition of prognosis in MLD patients, which is particularly relevant for pre-symptomatic diagnosis.


Subject(s)
Cerebroside-Sulfatase/genetics , Leukodystrophy, Metachromatic/genetics , Mutation , Animals , Genotype , HeLa Cells , Humans , Leukodystrophy, Metachromatic/enzymology , Mice , Mice, Knockout , Phenotype , Polymerase Chain Reaction
10.
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
11.
Acta Biomater ; 2(2): 221-7, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16701881

ABSTRACT

Encapsulation of pancreatic islets in alginate is used to protect against xenogenic rejection in different animal models. In this study, several factors, including differences in alginate composition, the presence or absence of xenogenic islet tissue and a transient immunosuppression, were investigated in a model of bovine islet transplantation in rats. A pure alginate with predominantly guluronic acid (Manugel) and an ultrapure low viscosity guluronic acid alginate (UP-LVG) were used. When microcapsules of Manugel or UP-LVG containing 16,000 bovine islet equivalents were transplanted in diabetic rats, we observed normoglycemia for 8.3+/-0.7 (range 6-12 days) and 7.5+/-0.2 days (range 7-8 days) on average, respectively. To ameliorate immunoprotection of alginate microcapsules we repeated the same experiments using transient immunosuppressive therapy. Low doses of cyclosporin A (CyA) administered for 18 days after implantation increased the time in normoglycemia, which averaged 27+/-3 days (range 8-55 days) in Manugel capsules while in UP-LVG capsules it averaged 18+/-8 days (range 3-39 days). The surface of recovered capsules showed less capsules free of overgrowth in Manugel with respect to UP-LVG alginate. These data were comparable with those observed in empty microcapsules similarly implanted, indicating that the capsular overgrowth was not promoted by the presence of xenogenic islet tissue. In recovered Manugel capsules the percentage of capsules without fibrotic overgrowth was higher than that observed without CyA. The same observation was made in empty capsules. These observations indicate that a combination of a highly purified alginate and short-term immunosuppression prolong islet function in a model of xenotransplantation.


Subject(s)
Capsules , Islets of Langerhans Transplantation/methods , Islets of Langerhans/cytology , Alginates , Animals , Biocompatible Materials , Blood Glucose/metabolism , Cattle , Cell Separation/methods , Centrifugation, Density Gradient , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/surgery , Glucuronic Acid , Hexuronic Acids , Male , Rats , Rats, Wistar , Transplantation, Heterologous , Ultracentrifugation
12.
Am J Pathol ; 168(1): 42-54, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16400008

ABSTRACT

Changes in podocyte number or density have been suggested to play an important role in renal disease progression. Here, we investigated the temporal relationship between glomerular podocyte number and development of proteinuria and glomerulosclerosis in the male Munich Wistar Fromter (MWF) rat. We also assessed whether changes in podocyte number affect podocyte function and focused specifically on the slit diaphragm-associated protein nephrin. Age-matched Wistar rats were used as controls. Estimation of podocyte number per glomerulus was determined by digital morphometry of WT1-positive cells. MWF rats developed moderate hypertension, massive proteinuria, and glomerulosclerosis with age. Glomerular hypertrophy was already observed at 10 weeks of age and progressively increased thereafter. By contrast, mean podocyte number per glomerulus was lower than normal in young animals and further decreased with time. As a consequence, the capillary tuft volume per podocyte was more than threefold increased in older rats. Electron microscopy showed important changes in podocyte structure of MWF rats, with expansion of podocyte bodies surrounding glomerular filtration membrane. Glomerular nephrin expression was markedly altered in MWF rats and inversely correlated with both podocyte loss and proteinuria. Our findings suggest that reduction in podocyte number is an important determinant of podocyte dysfunction and progressive impairment of the glomerular permselectivity that lead to the development of massive proteinuria and ultimately to renal scarring.


Subject(s)
Glomerulosclerosis, Focal Segmental/physiopathology , Membrane Proteins/metabolism , Podocytes/metabolism , Podocytes/ultrastructure , Animals , Blotting, Western , Disease Models, Animal , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Hypertension/etiology , Immunohistochemistry , Male , Microscopy, Electron, Scanning , Microscopy, Electron, Transmission , Proteinuria/etiology , RNA, Messenger/analysis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
13.
Biomaterials ; 26(28): 5640-7, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15878369

ABSTRACT

Transplantation of pancreatic islets in diabetes is currently limited by the need of immunosuppressive therapy. The present study was designed to test an immunoprotection planar device for subcutaneous xenotransplantation of pancreatic islets in the diabetic rat. We tested three different devices made of polyethersulfone hollow fibers. In all diabetic rats, implantation of islet-containing devices promptly normalized hyperglycemia. In vitro membrane permeability to glucose was correlated with implant function duration. These data confirm that bovine islets contained within devices and implanted subcutaneously remain functional for several days. Strategies to prolong islet function may allow achieving successful long-term islet implantation in this attractive site.


Subject(s)
Bioprosthesis , Cell Culture Techniques/instrumentation , Diabetes Mellitus/pathology , Diabetes Mellitus/surgery , Islets of Langerhans Transplantation/instrumentation , Islets of Langerhans/pathology , Pancreas, Artificial , Animals , Biocompatible Materials/chemistry , Blood Glucose/analysis , Cattle , Cell Culture Techniques/methods , Dermatologic Surgical Procedures , Diabetes Mellitus/blood , Equipment Failure Analysis , Feasibility Studies , Islets of Langerhans Transplantation/methods , Male , Materials Testing , Polymers/chemistry , Prosthesis Design , Rats , Rats, Wistar , Sulfones/chemistry , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...