Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Expert Opin Biol Ther ; 23(4): 353-364, 2023 04.
Article in English | MEDLINE | ID: mdl-36920351

ABSTRACT

INTRODUCTION: Lysosomal storage disorders (LSD) are a group of monogenic rare diseases caused by pathogenic variants in genes that encode proteins related to lysosomal function. These disorders are good candidates for gene therapy for different reasons: they are monogenic, most of lysosomal proteins are enzymes that can be secreted and cross-correct neighboring cells, and small quantities of these proteins are able to produce clinical benefits in many cases. Ex vivo gene therapy allows for autologous transplant of modified cells from different sources, including stem cells and hematopoietic precursors. AREAS COVERED: Here, we summarize the main gene therapy and genome editing strategies that are currently being used as ex vivo gene therapy approaches for lysosomal disorders, highlighting important characteristics, such as vectors used, strategies, types of cells that are modified and main results in different disorders. EXPERT OPINION: Clinical trials are already ongoing, and soon approved therapies for LSD based on ex vivo gene therapy approaches should reach the market.


Subject(s)
Lysosomal Storage Diseases , Humans , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/therapy , Genetic Vectors , Genetic Therapy/methods , Lysosomes
2.
PLoS One ; 10(6): e0128922, 2015.
Article in English | MEDLINE | ID: mdl-26067671

ABSTRACT

Glioblastoma (GBM) is an infiltrative tumor that is difficult to eradicate. Treating GBM with mesenchymal stem cells (MSCs) that have been modified with the HSV-Tk suicide gene has brought significant advances mainly because MSCs are chemoattracted to GBM and kill tumor cells via a bystander effect. To use this strategy, abundantly present adipose-tissue-derived mesenchymal stem cells (AT-MSCs) were evaluated for the treatment of GBM in mice. AT-MSCs were prepared using a mechanical protocol to avoid contamination with animal protein and transduced with HSV-Tk via a lentiviral vector. The U-87 glioblastoma cells cultured with AT-MSC-HSV-Tk died in the presence of 25 or 50 µM ganciclovir (GCV). U-87 glioblastoma cells injected into the brains of nude mice generated tumors larger than 3.5 mm2 after 4 weeks, but the injection of AT-MSC-HSV-Tk cells one week after the U-87 injection, combined with GCV treatment, drastically reduced tumors to smaller than 0.5 mm2. Immunohistochemical analysis of the tumors showed the presence of AT-MSC-HSV-Tk cells only within the tumor and its vicinity, but not in other areas of the brain, showing chemoattraction between them. The abundance of AT-MSCs and the easier to obtain them mechanically are strong advantages when compared to using MSCs from other tissues.


Subject(s)
Adipose Tissue/metabolism , Glioblastoma/metabolism , Mesenchymal Stem Cells/enzymology , Simplexvirus/genetics , Thymidine Kinase/biosynthesis , Transduction, Genetic , Viral Proteins/biosynthesis , Adipose Tissue/pathology , Animals , Bystander Effect/drug effects , Cell Line, Tumor , Female , Ganciclovir/pharmacology , Glioblastoma/pathology , Glioblastoma/therapy , Humans , Mesenchymal Stem Cells/pathology , Mice , Mice, Nude , Simplexvirus/enzymology , Thymidine Kinase/genetics , Viral Proteins/genetics
3.
J Gene Med ; 17(1-2): 1-13, 2015.
Article in English | MEDLINE | ID: mdl-25597593

ABSTRACT

BACKGROUND: Mucopolysaccharidose type I (MPSI) is a lysosomal monogenic disease caused by mutations in the gene for α- L-iduronidase (IDUA). MPSI patients need a constant supply of IDUA to alleviate progression of the disease. IDUA gene transfer using integrative vectors might provide a definitive solution and support advancement to clinical trials, although studies have not yet been satisfactory. To achieve a stable IDUA gene expression in vivo, phiC31 was tested in the present study. METHODS: Several plasmid vectors were constructed and IDUA-/- mice were treated with cyclophosphamide and transfected with these vectors hydrodynamically via tail veins. IDUA expression was monitored over time. Treated and nontreated mice underwent an open-field test at age 8 months, and IDUA activity and glycosaminoglycan (GAG) content of tissues were evaluated. RESULTS: High levels of IDUA activity were detected initially (>1000 U/ml), although these levels decayed over time. The reinjection of vectors produced a similar profile of IDUA decay. Three out of six treated mice had IDUA activity in the livers, and also showed lower GAG content, reduced lysosomes and better locomotion. To investigate unsustained IDUA production, wild-type mice were submitted to the same gene therapy procedure, which generated a similar profile of IDUA decay. Anti-IDUA antibody was detected in the sera of these animals. In addition, we also found three methylated sites in the cytomegalovirus promoter region. CONCLUSIONS: phiC31-mediated gene therapy resulted in an important improvement in IDUA-/- mice, including locomotion, although the obstacles that need to be overcome to enable long-term gene therapy for MPSI are also noted.


Subject(s)
Gene Transfer Techniques , Genetic Therapy , Genetic Vectors/genetics , Iduronidase/genetics , Mucopolysaccharidosis I/genetics , Animals , Behavior, Animal , Cell Line , DNA Methylation , Disease Models, Animal , Enzyme Activation , Female , Gene Expression , Gene Order , Genes, Reporter , Genetic Vectors/administration & dosage , HEK293 Cells , Homologous Recombination , Humans , Iduronidase/metabolism , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Knockout , Motor Activity , Mucopolysaccharidosis I/metabolism , Mucopolysaccharidosis I/therapy , Nucleotide Motifs , Promoter Regions, Genetic , Transfection
4.
Methods Mol Biol ; 1213: 147-69, 2014.
Article in English | MEDLINE | ID: mdl-25173381

ABSTRACT

Mesenchymal stem cells (MSCs) can be obtained from adult bone marrow and adipose tissue in large quantities and are the main cell types that contribute to recovery from ischemia because, among their biological activities, they produce several proangiogenic paracrine factors and differentiate into endothelial cells. Mouse hind limb ischemia induced by surgery is a useful animal model to study the angiogenic properties of MSCs, but it requires several precautions to be reproducible. The preparation of MSCs, the ischemic surgery, and the physiological and histological analyses are described in detail.


Subject(s)
Extremities/blood supply , Extremities/pathology , Ischemia/pathology , Ischemia/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Neovascularization, Physiologic , Adipogenesis , Animals , Cell Culture Techniques , Cell Differentiation , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/metabolism , Immunohistochemistry , Male , Mesenchymal Stem Cells/metabolism , Mice , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Osteogenesis
5.
PLoS One ; 9(3): e92420, 2014.
Article in English | MEDLINE | ID: mdl-24642723

ABSTRACT

Mucopolysaccharidosis type I (MPSI) is an autosomal recessive disease that leads to systemic lysosomal storage, which is caused by the absence of α-L-iduronidase (IDUA). Enzyme replacement therapy is recognized as the best therapeutic option for MPSI; however, high titers of anti-IDUA antibody have frequently been observed. Due to the immunosuppressant properties of MSC, we hypothesized that MSC modified with the IDUA gene would be able to produce IDUA for a long period of time. Sleeping Beauty transposon vectors were used to modify MSC because these are basically less-immunogenic plasmids. For cell transplantation, 4×10(6) MSC-KO-IDUA cells (MSC from KO mice modified with IDUA) were injected into the peritoneum of KO-mice three times over intervals of more than one month. The total IDUA activities from MSC-KO-IDUA before cell transplantation were 9.6, 120 and 179 U for the first, second and third injections, respectively. Only after the second cell transplantation, more than one unit of IDUA activity was detected in the blood of 3 mice for 2 days. After the third cell transplantation, a high titer of anti-IDUA antibody was detected in all of the treated mice. Anti-IDUA antibody response was also detected in C57Bl/6 mice treated with MSC-WT-IDUA. The antibody titers were high and comparable to mice that were immunized by electroporation. MSC-transplanted mice had high levels of TNF-alpha and infiltrates in the renal glomeruli. The spreading of the transplanted MSC into the peritoneum of other organs was confirmed after injection of 111In-labeled MSC. In conclusion, the antibody response against IDUA could not be avoided by MSC. On the contrary, these cells worked as an adjuvant that favored IDUA immunization. Therefore, the humoral immunosuppressant property of MSC is questionable and indicates the danger of using MSC as a source for the production of exogenous proteins to treat monogenic diseases.


Subject(s)
Iduronidase/immunology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Mucopolysaccharidosis I/therapy , Animals , Autoantibodies/blood , Cells, Cultured , Combined Modality Therapy , Cytokines/blood , Enzyme Replacement Therapy , Humans , Iduronidase/therapeutic use , Injections, Intraperitoneal , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucopolysaccharidosis I/blood , Mucopolysaccharidosis I/immunology , Tissue Distribution
6.
Stem Cell Res Ther ; 4(4): 86, 2013 Jul 26.
Article in English | MEDLINE | ID: mdl-23890057

ABSTRACT

INTRODUCTION: BALB/c mice and C57/BL6 mice have different abilities to recover from ischemia. C57/BL6 mice display increased vessel collateralization and vascular endothelial growth factor expression with a consequent rapid recovery from ischemia compared with BALB/c mice. Mesenchymal stem cells (MSCs) are one of the main cell types that contribute to the recovery from ischemia because, among their biological activities, they produce several proangiogenic paracrine factors and differentiate into endothelial cells. The objective of this study was to evaluate whether the MSCs of these two mouse strains have different inductive capacities for recovering ischemic limbs. METHODS: MSCs from these two strains were obtained from the bone marrow, purified and characterized before being used for in vivo experiments. Limb ischemia was surgically induced in BALB/c mice, and MSCs were injected on the fifth day. The evolution of limb necrosis was evaluated over the subsequent month. Muscle strength was assessed on the 30th day after the injection, and then the animals were sacrificed to determine the muscle mass and perform histological analyses to detect cellular infiltration, capillary and microvessel densities, fibrosis, necrosis and tissue regeneration. RESULTS: The MSCs from both strains promoted high level of angiogenesis similarly, resulting in good recovery from ischemia. However, BALB/c MSCs promoted more muscle regeneration (57%) than C57/BL6 MSCs (44%), which was reflected in the increased muscle strength (0.79 N versus 0.45 N). CONCLUSION: The different genetic background of MSCs from BALB/c mice and C57/BL6 mice was not a relevant factor in promoting angiogenesis of limb ischemia, because both cells showed a similar angiogenic activity. These cells also showed a potential myogenic effect, but the stronger effect promoted by BALB/c MSCs indicates that the different genetic background of MSCs was more relevant in myogenesis than angiogenesis.


Subject(s)
Endothelial Cells/metabolism , Hindlimb/blood supply , Ischemia/metabolism , Animals , Bone Marrow , Cell- and Tissue-Based Therapy , Disease Models, Animal , Hindlimb/pathology , Ischemia/pathology , Male , Mesenchymal Stem Cells , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neovascularization, Physiologic
7.
Cytotherapy ; 15(7): 820-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23660332

ABSTRACT

BACKGROUND AIMS: Granulocyte macrophage-colony stimulating factor (GM-CSF) promotes vessel formation through several molecular signaling pathways. Mesenchymal stromal cells (MSCs) have an important role in neovasculogenesis during ischemia because they release pro-angiogenic paracrine factors, pro-survival and immunomodulatory substances and can differentiate into endothelial cells. The objective of this study was to evaluate whether there is synergy between GM-CSF and MSCs in recovering ischemic limbs. METHODS: MSCs from mouse bone marrow were transduced with a lentiviral vector expressing GM-CSF and injected into animals with surgically induced limb ischemia, with unmodified MCSs used as control. The evolution of limb necrosis was evaluated for 1 month. Muscle strength was assessed on the 30th day, and the animals were euthanized to determine the muscle mass and to perform histological analyses to determine the degree of cellular infiltration, capillary and microvessel densities, fibrosis, necrosis and tissue regeneration. RESULTS: Both treatments were able to ameliorate ischemia, decrease the areas of fibrosis, necrosis, adipocytes and leukocyte infiltrates and increase the number of capillaries. The addition of GM-CSF promoted the formation of larger vessels, but it also resulted in more fibrosis and less muscle mass without affecting muscle force. CONCLUSIONS: Both treatments resulted in a remarkable amelioration of ischemia. More fibrosis and less muscle mass produced by the overexpression of GM-CSF did not affect muscle functionality significantly. Importantly, MSCs overexpressing GM-CSF produced larger vessels, which is an important long-term advantage because larger vessels are more efficient in the reperfusion of ischemic tissues physiologically.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Peripheral Arterial Disease/therapy , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Extremities/blood supply , Extremities/pathology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Ischemia/pathology , Ischemia/therapy , Mesenchymal Stem Cells/metabolism , Mice , Necrosis/pathology , Necrosis/therapy , Peripheral Arterial Disease/pathology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...