1
|
Fenjves ES, Gordon DA, Pershing LK, Williams DL, Taichman LB. Systemic distribution of apolipoprotein E secreted by grafts of epidermal keratinocytes: implications for epidermal function and gene therapy. Proc Natl Acad Sci U S A 1989; 86:8803-7. [PMID: 2813425 PMCID: PMC298378 DOI: 10.1073/pnas.86.22.8803] [Citation(s) in RCA: 102] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
In the present study, human apolipoprotein E (apoE) was monitored in the circulation of athymic mice and rats bearing human epidermal grafts. Human apoE was detected in the systemic circulation of graft-bearing animals as long as the graft remained on the animal. Within 24 hr of graft removal, human apoE was not detectable in plasma, indicating that apoE resulted from continuous production of the protein by grafted keratinocytes. These results show that proteins as large as apoE (299 amino acids) traverse the epidermal-dermal barrier and achieve systemic distribution where they may produce effects on distal tissues. The feasibility of using grafts of genetically-altered keratinocytes for the delivery of secreted proteins is clearly reinforced by the demonstration that an epidermally derived protein exhibits systemic distribution. Finally, by virtue of its systemic distribution, apoE produced in a peripheral tissue such as skin, may function in the reverse transport of cholesterol from peripheral tissues to the liver.
Collapse
|
research-article |
36 |
102 |
2
|
Fenjves ES, Yao SN, Kurachi K, Taichman LB. Loss of expression of a retrovirus-transduced gene in human keratinocytes. J Invest Dermatol 1996; 106:576-8. [PMID: 8648197 DOI: 10.1111/1523-1747.ep12344976] [Citation(s) in RCA: 81] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Retroviral-mediated transfer of new genetic information into keratinocytes is a key step in epidermal gene therapy. An obstacle to the use of retroviruses for gene therapy is that although high levels of expression of the transduced gene can be maintained in tissue culture, expression is often lost when the cells are transplanted to an animal host. To examine some of the factors involved in this instability of expression, we transduced keratinocytes with a retrovirus encoding the gene for human factor IX and monitored secretion of the transduced gene. We observed continued secretion of factor IX through five passages in culture. When, however, sheets of these cells were grafted to athymic mice, factor IX expression was reduced or lost within 6 wk. We show that the reduction of factor IX expression in grafted keratinocytes did not result from a loss of grafted cells, nor was there a block to systemic delivery of a secreted endogenous product.
Collapse
|
|
29 |
81 |
3
|
Fenjves ES, Smith J, Zaradic S, Taichman LB. Systemic delivery of secreted protein by grafts of epidermal keratinocytes: prospects for keratinocyte gene therapy. Hum Gene Ther 1994; 5:1241-8. [PMID: 7849096 DOI: 10.1089/hum.1994.5.10-1241] [Citation(s) in RCA: 77] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Grafts of autologous keratinocytes genetically altered to secrete a new gene product are a potential vehicle for gene therapy. To consider the feasibility of such an approach, we have examined the ability of keratinocytes to secrete and deliver apolipoprotein E (apoE) to the circulation of mice bearing grafts of human keratinocytes. The grafted keratinocytes secreted two forms of apoE, an endogenous apoE encoded in the genome and a recombinant apoE encoded in a transfected gene construct. In vitro studies showed that endogenous apoE was secreted from basal keratinocytes whereas recombinant apoE was secreted from basal as well as suprabasal cells. On the basis of amounts of recombinant apoE present in the serum of grafted mice, we estimate that a graft occupying 2% of the surface area of an adult human would deliver 6.5-8.3 mg of recombinant apoE protein per day.
Collapse
|
|
31 |
77 |
4
|
Garlick JA, Katz AB, Fenjves ES, Taichman LB. Retrovirus-mediated transduction of cultured epidermal keratinocytes. J Invest Dermatol 1991; 97:824-9. [PMID: 1919048 DOI: 10.1111/1523-1747.ep12489019] [Citation(s) in RCA: 63] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Retrovirus-mediated gene transfer is an efficient means of introducing and expressing exogenous gene(s) in many cell types including keratinocytes. However, parameters of transduction and gene expression have not been systematically analyzed for keratinocytes. To carry out such a study we have transduced cultures of newborn foreskin cells with retroviral vectors that encode the genes for neomycin resistance (neor) and for beta-galactosidase (B-gal). The neor gene is a dominant selectable marker and the B-gal gene encodes a histochemically detectable product. Our key findings are the following: 1) all keratinocytes that form colonies can be successfully transduced at a viral titer greater than 5 x 10(6) colony-forming units/ml; 2) transduction is effected by integration of a single copy of retroviral DNA; 3) transduced cells are not at a growth disadvantage and, in fact, single clones of transduced keratinocytes can be expanded to yield over 10(9) cells, suggesting that stem cells are transduced; 4) whereas most transduced colonies exhibit B-gal staining in a high percentage of constituent cells, some colonies had a mosaic or sectored staining pattern; 5) expression of the non-selectable B-gal gene was somewhat greater in differentiated cells of the culture as compared to nondifferentiated precursors. The ability to transduce stem cells at a high efficiency and to follow expression of transduced genes in clonal progeny will allow lineage mapping in stratified epithelial tissues.
Collapse
|
|
34 |
63 |
5
|
Flotte T, Agarwal A, Wang J, Song S, Fenjves ES, Inverardi L, Chesnut K, Afione S, Loiler S, Wasserfall C, Kapturczak M, Ellis T, Nick H, Atkinson M. Efficient ex vivo transduction of pancreatic islet cells with recombinant adeno-associated virus vectors. Diabetes 2001; 50:515-20. [PMID: 11246870 DOI: 10.2337/diabetes.50.3.515] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The ability to transfer immunoregulatory, cytoprotective, or antiapoptotic genes into pancreatic islet cells may allow enhanced posttransplantation survival of islet allografts and inhibition of recurrent autoimmune destruction of these cells in type 1 diabetes. However, transient transgene expression and the tendency to induce host inflammatory responses have limited previous gene delivery studies using viral transfer vectors. We demonstrate here that recombinant adeno-associated virus (rAAV) serotype 2, a vector that can overcome these limitations, effectively transduces both human and murine pancreatic islet cells with reporter genes as well as potentially important immunoregulatory cytokine genes (interleukin-4, interleukin-10), although a very high multiplicity of infection (10,000 infectious units/islet equivalent) was required. This requirement was alleviated by switching to rAAV serotype 5, which efficiently transduced islets at a multiplicity of infection of 100. Although adenovirus (Ad) coinfection was required for efficient ex vivo expression at early time points, islets transduced without Ad expressed efficiently when they were transplanted under the renal capsule and allowed to survive in vivo. The rAAV-delivered transgenes did not interfere with islet cell insulin production and were expressed in both beta- and non-beta-cells. We believe rAAV will provide a useful tool to deliver therapeutic genes for modulating immune responses against islet cells and markedly enhance longterm graft survival.
Collapse
|
|
24 |
60 |
6
|
Fenjves ES, Ochoa MS, Cabrera O, Mendez AJ, Kenyon NS, Inverardi L, Ricordi C. Human, nonhuman primate, and rat pancreatic islets express erythropoietin receptors. Transplantation 2003; 75:1356-60. [PMID: 12717230 DOI: 10.1097/01.tp.0000062862.88375.bd] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Erythropoietin (EPO) promotes survival in a variety of cells by mediating antiapoptotic signals through the EPO receptor (R). The authors examined pancreatic islets for the presence of EPO-R to determine whether these cells are protected by EPO from cytokine-induced apoptosis. METHODS Reverse-transcriptase polymerase chain reaction, immunohistology, and Western blots were used to establish the presence and localization of EPO-R on rat, nonhuman primate, and human islets. Islets were exposed to cytokines in the presence and absence of recombinant EPO and apoptosis was measured using a terminal deoxynucleotide transferase-mediated dUTP nick-end labeling assay followed by fluorescence-activated cell sorter analysis. Glucose stimulation indices were measured to assess the effect of EPO on islet function. RESULTS The presence of EPO-R was demonstrated on islets regardless of species. Recombinant EPO protected islets in culture from cytokine-induced apoptosis in a dose-dependent manner. Furthermore, the presence of EPO in the media does not adversely affect islet function. CONCLUSIONS This is the first demonstration that pancreatic islets express EPO-R and that EPO may prevent islet-cell apoptosis in culture. In vivo trials to evaluate the potential of long-term expression of EPO to augment islet survival in transplantation are underway.
Collapse
|
|
22 |
55 |
7
|
Gordon DA, Fenjves ES, Williams DL, Taichman LB. Synthesis and secretion of apolipoprotein E by cultured human keratinocytes. J Invest Dermatol 1989; 92:96-9. [PMID: 2462600 DOI: 10.1111/1523-1747.ep13071302] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Non-polar lipids are synthesized by keratinocytes in the epidermis and transported to the extracellular space where they contribute to formation of a permeability barrier. Transport of non-polar lipids in other organs and tissues usually occurs with the lipid complexed to an apolipoprotein. In this study we set out to learn if apolipoprotein E is produced by human epidermal keratinocytes in culture. Analysis of total cellular RNA from cultured keratinocytes showed the presence of human apolipoprotein E mRNA at concentrations ranging from 2.5 to 35 molecules/cell. The cells secrete a protein identified as apo E on the basis of molecular weight, isoform pattern, and immunoreactivity. Enzyme linked immunosorbent assay of media from keratinocyte cultures indicated that apolipoprotein E is secreted at a rate of 0.92 ng/h/10(6) cells.
Collapse
|
|
36 |
40 |
8
|
Fenjves ES, Ochoa MS, Gay-Rabinstein C, Molano RD, Pileggi A, Mendez AJ, Inverardi L, Ricordi C. Adenoviral gene transfer of erythropoietin confers cytoprotection to isolated pancreatic islets. Transplantation 2004; 77:13-8. [PMID: 14724429 DOI: 10.1097/01.tp.0000110422.27977.26] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
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.
Collapse
|
Research Support, Non-U.S. Gov't |
21 |
30 |
9
|
Barra RM, Fenjves ES, Taichman LB. Secretion of apolipoprotein E by basal cells in cultures of epidermal keratinocytes. J Invest Dermatol 1994; 102:61-6. [PMID: 8288912 DOI: 10.1111/1523-1747.ep12371733] [Citation(s) in RCA: 29] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Recently it has been shown that apolipoprotein E (apoE) secreted by keratinocytes in transplanted epidermal grafts reaches the systemic circulation. In this study we ask which cells in cultures of epidermal keratinocytes, basal or suprabasal, are the source of apoE. By fractionating disaggregated cultures in gradients of Ficoll400, the small nondifferentiated cells derived from the basal compartment were shown to be the source of apoE. The larger more differentiated cells derived from suprabasal layers could not be shown to contain or secrete apoE, although they did contain the apoE mRNA. Basal cells are the primary site for replication. However, analysis during growth in culture indicated that secretion did not correlate with cell replication but appeared to be linked to specific changes in metabolic activity of the basal cell compartment. Localization of apoE secretion to the basal compartment may provide a mechanism for lipid uptake and redistribution within the epidermis and may be viewed within the larger context of keratinocyte differentiation.
Collapse
|
|
31 |
29 |
10
|
Fenjves ES, Schwartz PM, Blaese RM, Taichman LB. Keratinocyte gene therapy for adenosine deaminase deficiency: a model approach for inherited metabolic disorders. Hum Gene Ther 1997; 8:911-7. [PMID: 9195213 DOI: 10.1089/hum.1997.8.8-911] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Disorders in which there is toxic buildup of circulating substrate may be treated by furnishing an enzyme reservoir capable of metabolically processing the excess substrate. The epidermal keratinocyte is a potential site for such a reservoir. In this study, we explore the capacity of genetically modified keratinocytes to metabolize extracellular substrate in a culture model that resembles in vivo epidermal architecture. Keratinocytes from adenosine deaminase (ADA)-deficient patients were transduced with a retroviral vector encoding the human ADA gene and the capacity of this tissue to deaminate deoxyadenosine (dAdo) in vitro was measured. The results show that at a substrate concentration of 10 microM, ADA-corrected keratinocytes deaminate dAdo at a rate of 0.38 nmol/min.10(6) cells. These results indicate that keratinocytes process extracellular substrate at rates that suggest complete substrate conversion in a single pass. This study provides a strong indication that the epidermis, the largest and most accessible tissue of the body, is a valuable site for designing clinically relevant gene therapies.
Collapse
|
|
28 |
29 |
11
|
Curran MA, Ochoa MS, Molano RD, Pileggi A, Inverardi L, Kenyon NS, Nolan GP, Ricordi C, Fenjves ES. Efficient transduction of pancreatic islets by feline immunodeficiency virus vectors1. Transplantation 2002; 74:299-306. [PMID: 12177606 DOI: 10.1097/00007890-200208150-00003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Pancreatic islets transplanted into immunocompetent diabetic subjects are rapidly lost to apoptotic or lytic death or both. Genetic engineering of islets before transplantation with protective genes may enhance their posttransplantation survival. Accomplishing this goal requires the development of a safe, efficient vector for islet gene delivery. METHODS The ability of feline immunodeficiency virus (FIV) vectors to transfer a green fluorescent protein (GFP) gene to NIT-1 cells and primary islets was measured and compared with murine leukemia virus (MLV) and human immunodeficiency virus (HIV) vectors. Islets were examined using confocal microscopy to determine the extent and pattern of infection. Toxicity of the procedure was assessed via measurement of glucose stimulation indices and by reversion of diabetic mice using either FIV-infected or control islet transplants. RESULTS FIV effectively transduces islets with no untoward effect on the insulin secretion capacity of the beta cells. When FIV, HIV, and MLV GFP vectors were standardized to the same 293 cell titer and used to infect NIT-1 cells or whole islets, the FIV transduced equal or greater numbers of cells relative to the HIV vector and significantly more than the MLV vector. Islets transduced with FIV GFP were transplanted in a murine model for diabetes and were shown to revert diabetes and express GFP 4 weeks after transduction and 3 weeks after transplantation. CONCLUSIONS FIV transduction is a nontoxic and efficient method to genetically modify pancreatic islets and may prove promising for delivering genes to augment islet survival after transplantation.
Collapse
|
Comparative Study |
23 |
23 |
12
|
Vizzardelli C, Molano RD, Pileggi A, Berney T, Cattan P, Fenjves ES, Peel A, Fraker C, Ricordi C, Inverardi L. Neonatal porcine pancreatic cell clusters as a potential source for transplantation in humans: characterization of proliferation, apoptosis, xenoantigen expression and gene delivery with recombinant AAV. Xenotransplantation 2002; 9:14-24. [PMID: 12005100 DOI: 10.1034/j.1399-3089.2002.0o128.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Neonatal porcine islets are characterized by reproducible isolation success and high yields, sizable advantages over adult islets. In this work we have analyzed selected phenotypic and functional characteristics of porcine neonatal islets relevant to their possible use for transplant in humans. We show that porcine islet cells proliferate in culture, and synthesize and store islet-specific hormones. Proliferating beta cells can be easily identified. Implant of cultured neonatal islets in immunodeficient rodents results in the reversal of diabetes, albeit with delay. We also show that measurable apoptosis occurs in cultured neonatal porcine islets. Further, antigens recognized by human natural antibodies are expressed in a dynamic fashion over the culture period analyzed and are not limited to the alpha-Gal epitope. Lastly, we demonstrate that a recombinant Adeno-Associated virus can be used to efficiently deliver a reporter gene in porcine islets. This characterization might be helpful in the definition of the potential use of neonatal porcine islets for human transplantation.
Collapse
|
|
23 |
23 |
13
|
Abstract
Type 1 diabetes mellitus is an autoimmune disorder in which the insulin-producing beta-cells of the pancreatic islets of Langerhans are selectively destroyed. Transplantation of allogeneic islets offers a novel therapeutic approach for type 1 diabetic patients. Primary obstacles to the successful outcome of this treatment are loss of the islets occurring first during the isolation procedure and then immediately following transplantation. The genetic make up of beta-cells contributes to making them particularly vulnerable to apoptosis and necrosis-induced cell death caused by the trauma of the isolation procedure and by non-specific inflammatory events at the transplantation site. In this review we present description of chemical and molecular biology based strategies to confer cytoprotection to beta-cells.
Collapse
|
Review |
20 |
16 |
14
|
Abstract
The introduction and expression of exogenous genetic material in cultured cells has provided a powerful tool for studying gene function and regulation. Immortalized cell lines have been useful for establishing gene transfer methodologies that are generally inefficient. For investigators of epidermal and mucosal biology, wishing to make use of the tissue architecture produced by primary keratinocytes in vitro, the limited life span of these cells presents a host of unique problems. Primary cells require the use of gene transfer methods that are highly efficient and will not significantly alter the cell's normal differentiation pathway. The purpose of this review is to evaluate gene transfer technology as it applies to keratinocytes.
Collapse
|
Review |
31 |
14 |
15
|
Garlick JA, Fenjves ES. Keratinocyte gene transfer and gene therapy. CRITICAL REVIEWS IN ORAL BIOLOGY AND MEDICINE : AN OFFICIAL PUBLICATION OF THE AMERICAN ASSOCIATION OF ORAL BIOLOGISTS 1996; 7:204-21. [PMID: 8909878 DOI: 10.1177/10454411960070030101] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Gene therapy has moved beyond the pre-clinical stage to the treatment of a variety of inherited and acquired diseases. For such therapy to be successful, genes must be efficiently delivered to target cells and gene products must be expressed for prolonged periods of time without toxic effects to the host. This may be achieved by means of an in vivo strategy where genes are transferred directly into a host cell, or by means of an ex vivo approach through which cells are removed, cultured, targeted for gene delivery, and grafted back to the host. Several obstacles continue to delay safe and effective clinical application of gene therapy in a variety of target cells. The limited survival of transplanted cells, transient expression of transferred genes, and difficulties in targeting stem cells are technical issues requiring further investigation. Epidermal and oral keratinocytes are potential vehicles for gene therapy. Several features of these tissues can be utilized to achieve delivery of therapeutic gene products for local or systemic delivery. These qualities include: (1) the presence of stem cells; (2) the cell-, strata-, and site-specific regulation of keratinocyte gene expression; (3) tissue accessibility; and (4) secretory capacity. Such features can be exploited by the use of gene therapy strategies to facilitate: (1) identification, enrichment, and targeting of stem cells to ensure the continued presence of the transferred gene; (2) high-level and persistent transgene expression using keratinocyte-specific promoters; (3) tissue access needed for culture and grafting for ex vivo therapy and direct in vivo gene transfer; (4) secretion of transgene product for local or systemic delivery; and (5) monitoring of genetically modified tissue and removal if treatment termination is required. Optimal gene therapy strategies are being tested in a variety of tissues to treat dominant and recessive genetic disorders as well as acquired diseases such as neoplasia and infectious disease. This experience provides a basis for the application of such clinical studies to a spectrum of diseases effecting epidermal and oral keratinocytes. Gene therapy is in an early stage yet holds great promise for its ultimate clinical application.
Collapse
|
Review |
29 |
13 |
16
|
Fenjves ES, Ochoa MS, Gay-Rabinstein C, Ricordi C, Curran MA. Retrovirally Transferred Genes Inhibit Apoptosis in an Insulin-Secreting Cell Line: Implications for Islet Transplantation. Cell Transplant 2017; 13:489-96. [PMID: 15565861 DOI: 10.3727/000000004783983710] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
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 β-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-α-induced apoptosis in these cells. These genes may be ideal candidates to augment islet survival following transplantation.
Collapse
|
|
8 |
7 |
17
|
Fenjves ES, Ochoa MS, Cechin S, Gay-Rabinstein C, Pérez-Alvarez I, Ichii H, Mendez A, Ricordi C, Curran MA. Protection of human pancreatic islets using a lentiviral vector expressing two genes: cFLIP and GFP. Cell Transplant 2008; 17:793-802. [PMID: 19044206 DOI: 10.3727/096368908786516828] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
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.
Collapse
|
Research Support, Non-U.S. Gov't |
17 |
6 |
18
|
|
|
37 |
4 |
19
|
Fenjves ES, Lee JI, Garlick JA, Gordon DA, Williams DL, Taichman LB. Prospects for epithelial gene therapy. BASIC LIFE SCIENCES 1990; 53:215-23. [PMID: 2282036 DOI: 10.1007/978-1-4613-0637-5_17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
|
35 |
4 |
20
|
|
|
25 |
2 |
21
|
Fenjves ES, Ochoa MS, Barney T, Molano D, Pileggi A, Ricordi C, Inverardi L. EVALUATION OF EX-VIVO GENE TRANSFER OF FADD-DN TO PANCREATIC ISLETS. ScientificWorldJournal 2001. [DOI: 10.1100/tsw.2001.23.207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
|
24 |
|
22
|
Fenjves ES, Ochoa MS, Barney T, Molano D, Pileggi A, Ricordi C, Inverardi L. Evaluation of Ex-Vivo Gene Transfer of Fadd-Dn to Pancreatic Islets. ScientificWorldJournal 2001; 1:92. [PMID: 30147564 PMCID: PMC6084190 DOI: 10.1100/tsw.2001.207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
|
24 |
|