Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Transplant ; 17(7): 793-802, 2008.
Article in English | MEDLINE | ID: mdl-19044206

ABSTRACT

Pancreatic islet transplantation can provide insulin independence to diabetic patients. However, apoptosis of islets often leads to early graft failure. Genetic engineering with protective gene(s) can improve the viability of these cells. Here we show successful transduction of human islets with a feline immunodeficiency virus (FIV) vector expressing both a cytoprotective (cFLIP) gene and the green fluorescent protein (GFP). Despite using low virus titers to maximize safety, transduced islets expressed both genes, resulting in improved beta-cell metabolic activity and viability. Although only approximately 10% of total islet cells were transduced, the significant viability advantages suggest a "barrier" effect in which protecting the periphery of the islet shields the core. These results provide the first demonstration that a lentiviral vector can express two genes in islets. Furthermore, the engineered islets are resistant to a variety of apoptotic stimuli, suggesting the potential of this approach in enhancing the viability of transplanted cells.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Gene Transfer Techniques , Genetic Vectors/genetics , Graft Rejection/prevention & control , Green Fluorescent Proteins/genetics , Immunodeficiency Virus, Feline/genetics , Islets of Langerhans/physiology , Antibiotics, Antineoplastic/metabolism , Apoptosis/physiology , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Cytokines/metabolism , Doxorubicin/metabolism , Genetic Vectors/metabolism , Glucose/metabolism , Green Fluorescent Proteins/metabolism , Humans , Immunodeficiency Virus, Feline/metabolism , Insulin/metabolism , fas Receptor/metabolism
2.
Transplantation ; 79(12): 1683-90, 2005 Jun 27.
Article in English | MEDLINE | ID: mdl-15973169

ABSTRACT

BACKGROUND: Almost half of all transplanted vascularized organ grafts will be lost to transplant arteriosclerosis sometime posttransplantation. Organ shortage for primary transplants and retransplants has led to donor-pool expansion to include elderly donors, knowing that aging per se promotes arteriosclerosis. The current understanding that donor age negatively affects organ and/or patient survival outcome is undermined by variables such as the use of immunosuppressive drugs, their toxicity to the graft, degree of donor-recipient histocompatibility, and the resulting chronic rejection. The purpose of this study was to determine whether the donor's age or recipient's age matters the most in transplant arteriosclerosis in the absence of such variables. METHODS: A syngeneic combination was used where young (2-month-old) and old (22-month-old) donor aortas were injured to initiate neointimal thickening, then transplanted into age-mismatched recipients for 14, 60, and 90 days and then assessed for neointimal thickening. Base level injury response due ischemia and surgery was evaluated in age-matched and noninjured aortic grafts, respectively. RESULTS: Young aortas invariably developed thicker neointima when transplanted into old recipients than when transplanted into young ones. Correspondingly, old aortas transplanted in young recipients consistently developed less neointimal thickening than when transplanted into old recipients. CONCLUSIONS: Our findings strongly suggest that the severity of age-related neointima formation is primarily determined by the recipient's age rather than the donor's age. Therefore, in addition to focusing on donor-specific tolerance induction, strategies aiming at increasing the lifespan of vascularized organ grafts also have to take into consideration the recipient's aging milieu.


Subject(s)
Aging/physiology , Aorta/transplantation , Arteriosclerosis/pathology , Transplantation, Homologous/pathology , Animals , Aorta/growth & development , Aorta/pathology , Aorta, Thoracic/pathology , Disease Models, Animal , Female , Graft Survival , Immunosuppressive Agents/therapeutic use , Rats , Rats, Inbred F344 , Transplantation, Homologous/immunology , Transplantation, Isogeneic , Tunica Intima/pathology
3.
Cell Transplant ; 13(5): 489-96, 2004.
Article in English | MEDLINE | ID: mdl-15565861

ABSTRACT

The transplantation of pancreatic islets for the treatment of type I diabetes is hindered by the enormous loss of cells due to early apoptotic events. Genetic engineering of islets with cytoprotective genes is an important strategy aimed to enhance the survival of these cells in the transplant setting. The present study was designed to evaluate and compare the effects of five genes on a cell line derived from insulin-producing beta-cells, NIT-1. Cells were transduced using a Maloney murine leukemia virus (MLV) vector coding for yellow fluorescent protein (YFP) and for one of the following antiapoptotic genes: cFLIP, FADD-DN, BcL-2, PI-9, and ICAM-2. These genes were able to protect NIT-1 cells from cytokine-induced apoptosis to varying degrees ranging from no protection to significant protection equivalent to an optimal dose of a chemical caspase inhibitor. The data demonstrate that cFLIP, FADD-DN, and PI-9 are significantly more effective in protecting NIT-1 cells than BcL-2 and ICAM-2. Additionally, the data show that despite its weak in vitro inhibition of caspase-3, PI-9 affords significant protection against TNF-alpha-induced apoptosis in these cells. These genes may be ideal candidates to augment islet survival following transplantation.


Subject(s)
Apoptosis , Gene Transfer Techniques , Insulin/metabolism , Retroviridae/genetics , Animals , Bacterial Proteins/genetics , Caspase 3 , Caspases/metabolism , Cell Line , Cell Survival , Cell Transplantation , Cytokines/metabolism , Genetic Engineering , Genetic Vectors , In Situ Nick-End Labeling , Insulinoma/metabolism , Islets of Langerhans Transplantation , Luminescent Proteins/genetics , Mice , Models, Biological , Moloney murine leukemia virus/genetics , Time Factors , Transgenes , Tumor Necrosis Factor-alpha/metabolism
4.
J Immunol ; 172(8): 4709-16, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-15067046

ABSTRACT

T cell diversity was once thought to depend on the interaction of T cell precursors with thymic epithelial cells. Recent evidence suggests, however, that diversity might arise through the interaction of developing T cells with other cells, the identity of which is not known. In this study we show that T cell diversity is driven by B cells and Ig. The TCR V beta diversity of thymocytes in mice that lack B cells and Ig is reduced to 6 x 10(2) from wild-type values of 1.1 x 10(8); in mice with oligoclonal B cells, the TCR V beta diversity of thymocytes is 0.01% that in wild-type mice. Adoptive transfer of diverse B cells or administration of polyclonal Ig increases thymocyte diversity in mice that lack B cells 8- and 7-fold, respectively, whereas adoptive transfer of monoclonal B cells or monoclonal Ig does not. These findings reveal a heretofore unrecognized and vital function of B cells and Ig for generation of T cell diversity and suggest a potential approach to immune reconstitution.


Subject(s)
B-Lymphocyte Subsets/immunology , Receptors, Antigen, T-Cell, alpha-beta/biosynthesis , Adoptive Transfer , Animals , Apoptosis/genetics , Apoptosis/immunology , B-Lymphocyte Subsets/pathology , B-Lymphocyte Subsets/transplantation , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Division/genetics , Cell Division/immunology , Cell Movement/genetics , Cell Movement/immunology , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Immunoglobulin G/administration & dosage , Immunoglobulin Heavy Chains/genetics , Immunoglobulin J-Chains/genetics , Lymphopenia/genetics , Lymphopenia/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Thymus Gland/immunology , Thymus Gland/metabolism , Thymus Gland/pathology
5.
Transplantation ; 77(1): 13-8, 2004 Jan 15.
Article in English | MEDLINE | ID: mdl-14724429

ABSTRACT

BACKGROUND: The transfer of cytoprotective genes to isolated pancreatic islets may contribute to their enhanced survival in the transplant setting. Our laboratory established the expression of functional erythropoietin (EPO) receptors throughout pancreatic islets. Because EPO is a cytokine that promotes survival, we examined whether adenovirus-mediated gene transfer of EPO would result in cytoprotection of human pancreatic islets in culture and in the transplant setting. METHODS: Isolated human islets were transduced using an adenoviral vector coding for human EPO or green fluorescent protein. Comparison of cell death in culture was measured using annexin V-phycoerythrin and propidium iodide. Transplantation of transduced islets into diabetic nude mice was used to assess the effect of EPO on islet function and in vivo survival. RESULTS: Adenoviral delivery of EPO to pancreatic islets resulted in high-level EPO synthesis and secretion, which did not affect islet function in vitro or in vivo. Islets transduced with EPO were protected from apoptosis in culture and were at a functional advantage in vivo when compared with islets transduced with green fluorescent protein or untransduced islets. The high level of EPO had a negative effect on the blood chemistry of the animals that underwent transplantation. CONCLUSIONS: Overexpression of EPO protects islets from destruction and does not compromise islet function. Genetic engineering with EPO may be a viable approach for improving islet survival and engraftment in the transplant setting, but regulation of the gene's expression will be an important prerequisite to this strategy.


Subject(s)
Adenoviridae/genetics , Cytoprotection , Erythropoietin/genetics , Gene Transfer Techniques , Genetic Vectors , Islets of Langerhans/cytology , Animals , Blood/drug effects , Erythropoietin/adverse effects , Erythropoietin/metabolism , Erythropoietin/pharmacology , Humans , In Vitro Techniques , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans Transplantation/adverse effects , Mice , Mice, Nude
SELECTION OF CITATIONS
SEARCH DETAIL
...