Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Med ; 16(5-6): 216-21, 2010.
Article in English | MEDLINE | ID: mdl-20454522

ABSTRACT

The protein transduction domain from human immunodeficiency virus (HIV) Tat allows proteins to penetrate the cell membrane. Enhanced cellular uptake of therapeutic proteins could benefit a number of disorders. This is especially true for lysosomal storage disorders (LSDs) where enzyme replacement therapy (ERT) and gene therapy have been developed. We developed a novel recombinant lentiviral vector (LV) that engineers expression of alpha-galactosidase A (alpha-gal A)-Tat fusion protein for correction of Fabry disease, the second-most prevalent LSD with manifestations in the brain, kidney and heart. In vitro experiments confirmed mannose-6-phosphate independent uptake of the fusion factor. Next, concentrated therapeutic LV was injected into neonatal Fabry mice. Analysis of tissues at 26 wks demonstrated similar alpha-gal A enzyme activities but enhanced globotriaosylceramide (Gb3) reduction in hearts and kidneys compared with the alpha-gal A LV control. This strategy might advance not only gene therapy for Fabry disease and other LSDs, but also ERT, especially for cardiac Fabry disease.


Subject(s)
Enzyme Replacement Therapy/methods , Fabry Disease/therapy , Genes, tat , Genetic Therapy/methods , alpha-Galactosidase/metabolism , Animals , Disease Models, Animal , Fabry Disease/genetics , Fabry Disease/metabolism , HIV/genetics , HeLa Cells , Humans , Kidney/enzymology , Kidney/metabolism , Lentivirus/genetics , Mice , Myocardium/enzymology , Myocardium/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transduction, Genetic , Trihexosylceramides/metabolism , alpha-Galactosidase/blood , alpha-Galactosidase/genetics
2.
Mol Ther ; 17(2): 262-8, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19002160

ABSTRACT

Myocardial infarction (MI) and subsequent adverse remodeling cause heart failure. Previously we demonstrated a role for Kit ligand (KL) in improving cardiac function post-MI. KL has two major isoforms; KL-1 is secreted whereas KL-2 is predominantly membrane bound. We demonstrate here first that KL-2-deficient mice have worse survival and an increased heart/bodyweight ratio post-MI compared to mice with reduced c-Kit receptor expression. Next we synthesized recombinant lentiviral vectors (LVs) that engineered functional expression of murine KL-1 and KL-2. For in vivo analyses, we directly injected these LVs into the left ventricle of membrane-bound KL-deficient Sl/Sl(d) or wild-type (WT) mice undergoing MI. Control LV/enGFP injection led to measurable reporter gene expression in hearts. Injection of LV/KL-2 attenuated adverse left ventricular remodeling and dramatically improved survival post-MI in both Sl/Sl(d) and WT mice (from 12 to 71% and 35 to 73%, respectively, versus controls). With regard toward beginning to understand the possible salutary mechanisms involved in this effect, differential staining patterns of Sca-1 and Ly49 on peripheral blood (PB) cells from therapeutically treated animals was found. Our data show that LV/KL-2 gene therapy is a promising treatment for MI.


Subject(s)
Injections/methods , Lentivirus/genetics , Myocardial Infarction/pathology , Myocardial Infarction/therapy , Stem Cell Factor/genetics , Animals , Cells, Cultured , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Mice , Myocardial Infarction/genetics , Myocardium/metabolism , Myocardium/pathology , Stem Cell Factor/physiology
3.
Mol Ther ; 16(3): 607-17, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18180774

ABSTRACT

Gene therapy strategies may accelerate the development of prophylactic immunotherapy against cancer. We synthesized a lentiviral (LV) vector encoding a kinase-deficient form of erbB2 (erbB2tr) to transduce murine dendritic cells (DCs) efficiently. Murine erbB2 models a clinically relevant tumor-associated self-antigen; its human homolog (HER-2/neu) is overexpressed in breast cancer and in 80% of metastatic prostate cancers. Following one infection, approximately 47% of DCs overexpressed erbB2tr. To determine whether low doses of transduced DCs could protect mice from prostate cancer cells, we performed prime/boost vaccinations with 2 x 10(3) or 2 x 10(5) erbB2tr-transduced DCs. Six weeks after vaccination, mice were simultaneously bilaterally challenged with the aggressive RM-1 prostate cancer cell line and an erbB2tr-expressing variant (RM-1-erbB2tr). Whereas control mice developed both tumors, all recipients of 2 x 10(5) erbB2tr-transduced DCs developed only wild-type RM-1 tumors. One-third of mice vaccinated with just 2 x 10(3) erbB2tr-transduced DCs also demonstrated erbB2tr-specific tumor protection. Protection against RM-1-erbB2tr tumors was associated with sustained levels of anti-erbB2tr antibody production and also correlated with erbB2tr-specific Th1 cytokine secretion. Depletion of CD4(+), CD8(+), or natural killer (NK) cells prior to tumor challenge underscored their role in mediating tumor protection. We conclude that administration of DCs expressing a self-antigen through efficient LV-based gene transfer activates cellular and humoral immunity, protecting host animals against specific tumor challenge.


Subject(s)
Dendritic Cells/immunology , Lentivirus/genetics , Prostatic Neoplasms/therapy , Receptor, ErbB-2/genetics , Vaccination/methods , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Line, Tumor , Cytokines/metabolism , Dendritic Cells/metabolism , Dendritic Cells/transplantation , Female , Flow Cytometry , Genetic Therapy/methods , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunotherapy, Adoptive/methods , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptor, ErbB-2/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Spleen/cytology , Spleen/metabolism , Transduction, Genetic
5.
Mol Ther ; 15(3): 618-27, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17228315

ABSTRACT

Hematopoietic cell transplantation can impact lysosomal storage disorders (LSDs) and will be enhanced by gene therapy. Transduced cells in LSDs often secrete the therapeutic hydrolase, which can be used by bystander cells. However, toxicity associated with myeloablative transplant preparative regimens limits many applications of this approach in gene therapy. We hypothesized that reduced-intensity (RI) conditioning regimens would allow stable engraftment of therapeutically transduced cells and allow correction of Fabry disease. We transplanted transduced cells into Fabry mice receiving eight different clinically relevant chemotherapy- and/or radiotherapy-based RI conditioning regimens generating modest and transient lymphoid/myeloid cell depletion. Two comprehensive transplantation Protocols were performed. Firstly, transplantation of 0.38 x 10(6) gene-modified stem/progenitor cells was nominally effective; none of the RI regimens led to stable alpha-galactosidase A (alpha-gal A) correction. Secondly, transduced cells were preselected for functional transgene expression and transplanted at a higher dose (0.72 x 10(6) cells). Each RI regimen yielded engraftment of functional transgene-positive cells through 180 days along with increased plasma alpha-gal A activity. Importantly, the RI regimens mediated broad organ enzyme correction and were not associated with immune responses against alpha-gal A. RI conditioning thus has an important role in gene therapy for LSDs; a variety of regimens can be effective in this context.


Subject(s)
Bone Marrow Transplantation/methods , Fabry Disease/therapy , Transplantation Conditioning/methods , Animals , Biomarkers , Fabry Disease/genetics , Fabry Disease/immunology , Fabry Disease/surgery , Gene Expression , Genetic Therapy , Humans , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Male , Mice , Spleen/metabolism
6.
Circ J ; 70(11): 1503-8, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17062978

ABSTRACT

BACKGROUND: Recombinant lentiviral vectors (LVs) offer the possibility of stable, long-term expression of transgenes even in non-dividing cells. In the present study this vector system was applied to a clinically relevant cardiovascular problem. METHODS AND RESULTS: Fabry disease results from deficient activity of alpha-galactosidase A (alpha-gal A) and cardiac abnormalities are a common and an important cause of death in patients with the disease. A therapeutic LV that delivers the alpha-gal A cDNA has been synthesized. In vitro studies established efficient transduction of the H9c2 rat cardiomyocytes and showed overexpression of enGFP (control) and alpha-gal A. In in vivo studies, the enGFP cDNA was transferred into C57BL/6 mouse hearts by direct intraventricular injection. Next, in a mouse model of Fabry disease, the recombinant therapeutic construct was delivered analogously. In cardiac tissue, alpha-gal A activity rose to 23% of normal levels at day 7 after LV injection, which is encouraging because levels of correction approximating 5% of normal may be curative for this disorder. There was also a corresponding reduction in globotriaosylceramide accumulation. Other organs assayed showed no detectable changes in alpha-gal A activity levels in injected animals. CONCLUSION: A localized benefit of directly injecting a therapeutic LV into the heart has been shown, confirming the utility of this delivery system for research and therapy for a variety of cardiovascular disorders.


Subject(s)
Fabry Disease/enzymology , Fabry Disease/therapy , Genetic Therapy/methods , Lentivirus/genetics , Myocytes, Cardiac/metabolism , alpha-Galactosidase/genetics , alpha-Galactosidase/metabolism , Animals , Cell Line , Disease Models, Animal , Fabry Disease/genetics , Fabry Disease/pathology , Gene Expression Regulation, Enzymologic , Genetic Vectors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Heart Ventricles , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/pathology , Rats , Transgenes/genetics
7.
FEBS J ; 273(9): 2064-75, 2006 May.
Article in English | MEDLINE | ID: mdl-16724420

ABSTRACT

We have shown that the ABC transporter, multiple drug resistance protein 1 (MDR1, P-glycoprotein) translocates glucosyl ceramide from the cytosolic to the luminal Golgi surface for neutral, but not acidic, glycosphingolipid (GSL) synthesis. Here we show that the MDR1 inhibitor, cyclosporin A (CsA) can deplete Gaucher lymphoid cell lines of accumulated glucosyl ceramide and Fabry cell lines of globotriaosyl ceramide (Gb3), by preventing de novo synthesis. In the Fabry mouse model, Gb3 is increased in the heart, liver, spleen, brain and kidney. The lack of renal glomerular Gb3 is retained, but the number of verotoxin 1 (VT1)-staining renal tubules, and VT1 tubular targeting in vivo, is markedly increased in Fabry mice. Adult Fabry mice were treated with alpha-galactosidase (enzyme-replacement therapy, ERT) to eliminate serum Gb3 and lower Gb3 levels in some tissues. Serum Gb3 was monitored using a VT1 ELISA during a post-ERT recovery phase +/- biweekly intra peritoneal CsA. After 9 weeks, tissue Gb3 content and localization were determined using VT1/TLC overlay and histochemistry. Serum Gb3 recovered to lower levels after CsA treatment. Gb3 was undetected in wild-type liver, and the levels of Gb3 (but not gangliosides) in Fabry mouse liver were significantly depleted by CsA treatment. VT1 liver histochemistry showed Gb3 accumulated in Kupffer cells, endothelial cell subsets within the central and portal vein and within the portal triad. Hepatic venule endothelial and Kupffer cell VT1 staining was considerably reduced by in vivo CsA treatment. We conclude that MDR1 inhibition warrants consideration as a novel adjunct treatment for neutral GSL storage diseases.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Cyclosporine/pharmacology , Fabry Disease/blood , Fabry Disease/drug therapy , Liver/metabolism , Trihexosylceramides/blood , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , Animals , Cell Line , Disease Models, Animal , Fabry Disease/enzymology , Fabry Disease/metabolism , Gaucher Disease/drug therapy , Kidney/chemistry , Kidney/drug effects , Liver/drug effects , Mice , Trihexosylceramides/metabolism , alpha-Galactosidase/therapeutic use
8.
Proc Natl Acad Sci U S A ; 101(48): 16909-14, 2004 Nov 30.
Article in English | MEDLINE | ID: mdl-15550536

ABSTRACT

Successful therapy for many inherited disorders could be improved if the intervention were initiated early. This is especially true for lysosomal storage disorders. Earlier intervention may allow metabolic correction to occur before lipid buildup has irreversible consequences and/or before the immune system mounts limiting responses. We have been developing gene therapy to treat lysosomal storage disorders, especially Fabry disease. We describe studies directed toward metabolic correction in neonatal animals mediated by recombinant lentiviral vectors. To develop this method, we first injected a marking lentiviral vector that engineers expression of luciferase into the temporal vein of recipient neonatal animals. The use of a cooled charged-coupled device camera allowed us to track transgene expression over time in live animals. We observed intense luciferase expression in many tissues, including the brain, that did not diminish over 24 weeks. Next, we injected neonatal Fabry mice a single time with a therapeutic lentiviral vector engineered to express human alpha-galactosidase A. The injection procedure was well tolerated. We observed increased plasma levels of alpha-galactosidase A activity starting at our first plasma collection point (4 weeks). Levels of alpha-galactosidase A activity were found to be significantly elevated in many tissues even after 28 weeks. No immune response was observed against the corrective transgene product. Increased levels of enzyme activity also led to significant reduction of globotriaosylceramide in the liver, spleen, and heart. This approach provides a method to treat lysosomal storage disorders and other disorders before destructive manifestations occur.


Subject(s)
Fabry Disease/therapy , Genetic Vectors , Lentivirus/genetics , Transgenes , Animals , Animals, Newborn , Cell Line , Chromatography, High Pressure Liquid , Enzyme-Linked Immunosorbent Assay , Fabry Disease/genetics , Humans , Immunohistochemistry , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...