1
|
Klinker MW, Reed TJ, Fox DA, Lundy SK. Interleukin-5 supports the expansion of fas ligand-expressing killer B cells that induce antigen-specific apoptosis of CD4(+) T cells and secrete interleukin-10. PLoS One 2013; 8:e70131. [PMID: 23940537 PMCID: PMC3734024 DOI: 10.1371/journal.pone.0070131] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 06/15/2013] [Indexed: 01/19/2023] Open
Abstract
Beyond their critical role in humoral immunity, B lymphocytes can employ a variety of immunomodulatory mechanisms including expression of the apoptosis-inducing molecule Fas ligand (FasL; CD178). Here, we extensively characterized the surface phenotype of FasL+ killer B cells, showing they are enriched in the IgMhighCD5+CD1dhigh B cell subset previously reported to contain a higher frequency of B cells producing interleukin-10 (IL-10). A rare population of B cells expressing IL-10 was present among FasL+ B cells, but most FasL+ B cells did not produce IL-10. We also identify interleukin-5 (IL-5) as a novel inducer of killer B cell function. Constitutively FasL+ B cells expressed higher levels of the IL-5 receptor, and treating B cells with IL-5 and CD40L resulted in the expansion of a B cell population enriched for FasL+ cells. B cells stimulated with IL-5 and CD40L were potent inducers of apoptosis in activated primary CD4+ T cells, and this killing function was antigen-specific and dependent upon FasL. IL-5 also enhanced IL-10 secretion in B cells stimulated with CD40L. Taken together these findings elucidate the relationship of FasL+ B cells and IL-10-producing B cells and demonstrate that IL-5 can induce or enhance both killer B cell activity and IL-10 secretion in B cells. Finally, we found that the killer B cell activity induced by IL-5 was completely blocked by IL-4, suggesting the existence of a previously unknown antagonistic relationship between these type-2 cytokines in modulating the activity of killer B cells. Targeting this IL-5/IL-4 signaling axis may therefore represent a novel area of drug discovery in inflammatory disorders.
Collapse
Affiliation(s)
- Matthew W. Klinker
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Tamra J. Reed
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - David A. Fox
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Steven K. Lundy
- Graduate Program in Immunology, University of Michigan, Ann Arbor, Michigan, United States of America
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
2
|
Macauley MS, Pfrengle F, Rademacher C, Nycholat CM, Gale AJ, von Drygalski A, Paulson JC. Antigenic liposomes displaying CD22 ligands induce antigen-specific B cell apoptosis. J Clin Invest 2013; 123:3074-83. [PMID: 23722906 DOI: 10.1172/jci69187] [Citation(s) in RCA: 180] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Accepted: 04/04/2013] [Indexed: 01/13/2023] Open
Abstract
Antibodies confer humoral immunity but can also be harmful when they target an autoantigen, alloantigen, allergen, or biotherapeutic. New strategies are needed for antigen-specific suppression of undesired antibody responses, particularly to T cell-dependent protein antigens, because they elicit T cell help. Here we show that liposomal nanoparticles, displaying both antigen and glycan ligands of the inhibitory coreceptor CD22, induce a tolerogenic program that selectively causes apoptosis in mouse and human B cells. These SIGLEC-engaging tolerance-inducing antigenic liposomes (STALs, where SIGLEC is defined as sialic acid-binding Ig-like lectin) induced robust antigen-specific tolerance to protein antigens in mice, preventing subsequent immune response to challenge with the same antigen. Since development of inhibitory antibodies to FVIII is a serious problem in treatment of hemophilia A patients, we investigated the potential of this approach for inducing tolerance to FVIII in a hemophilia mouse model. STALs prevented formation of inhibitory FVIII antibodies, allowing for effective administration of FVIII to hemophilia mice to prevent bleeding. These findings suggest that STALs could be used to eliminate or prevent harmful B cell-mediated immune responses.
Collapse
Affiliation(s)
- Matthew S Macauley
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA
| | | | | | | | | | | | | |
Collapse
|
3
|
Specific immunotherapy of experimental myasthenia gravis in vitro and in vivo: The Guided Missile strategy. J Neuroimmunol 2012; 251:25-32. [DOI: 10.1016/j.jneuroim.2012.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Revised: 06/05/2012] [Accepted: 06/11/2012] [Indexed: 11/21/2022]
|
4
|
Pro- and anti-apoptotic CD95 signaling in T cells. Cell Commun Signal 2011; 9:7. [PMID: 21477291 PMCID: PMC3090738 DOI: 10.1186/1478-811x-9-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Accepted: 04/08/2011] [Indexed: 12/20/2022] Open
Abstract
The TNF receptor superfamily member CD95 (Fas, APO-1, TNFRSF6) is known as the prototypic death receptor in and outside the immune system. In fact, many mechanisms involved in apoptotic signaling cascades were solved by addressing consequences and pathways initiated by CD95 ligation in activated T cells or other "CD95-sensitive" cell populations. As an example, the binding of the inducible CD95 ligand (CD95L) to CD95 on activated T lymphocytes results in apoptotic cell death. This activation-induced cell death was implicated in the control of immune cell homeostasis and immune response termination. Over the past years, however, it became evident that CD95 acts as a dual function receptor that also exerts anti-apoptotic effects depending on the cellular context. Early observations of a potential non-apoptotic role of CD95 in the growth control of resting T cells were recently reconsidered and revealed quite unexpected findings regarding the costimulatory capacity of CD95 for primary T cell activation. It turned out that CD95 engagement modulates TCR/CD3-driven signal initiation in a dose-dependent manner. High doses of immobilized CD95 agonists or cellular CD95L almost completely silence T cells by blocking early TCR-induced signaling events. In contrast, under otherwise unchanged conditions, lower amounts of the same agonists dramatically augment TCR/CD3-driven activation and proliferation. In the present overview, we summarize these recent findings with a focus on the costimulatory capacity of CD95 in primary T cells and discuss potential implications for the T cell compartment and the interplay between T cells and CD95L-expressing cells including antigen-presenting cells.
Collapse
|
5
|
Schütz C, Hoves S, Halbritter D, Zhang HG, Mountz JD, Fleck M. Alloantigen specific deletion of primary human T cells by Fas ligand (CD95L)-transduced monocyte-derived killer-dendritic cells. Immunology 2011; 133:115-22. [PMID: 21342185 DOI: 10.1111/j.1365-2567.2011.03417.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Numerous studies have been performed in vitro and in various animal models to modulate the interaction of dendritic cells (DC) and T cells by Fas (CD95/Apo-1) signalling to delete activated T cells via induction of activation-induced cell death (AICD). Previously, we could demonstrate that Fas ligand (FasL/CD95L)-expressing 'killer-antigen-presenting cells' can be generated from human monocyte-derived mature DC (mDC) using adenoviral gene transfer. To evaluate whether these FasL-expressing mDC (mDC-FasL) could eliminate alloreactive primary human T cells in vitro, co-culture experiments were performed. Proliferation of human T cells was markedly reduced in primary co-cultures with allogeneic mDC-FasL, whereas a strong proliferative T-cell response could be observed in co-cultures with enhanced green fluorescent protein-transduced mDC. Inhibition of T-cell proliferation was related to the transduction efficiency, and the numbers of mDC-FasL present in co-cultures. In addition, proliferation of pre-activated alloreactive CD4(+) and CD8(+) T cells could be almost completely inhibited in secondary co-cultures using mDC-FasL as stimulatory cells, which was the result of induction of apoptosis in the majority of preactivated T cells. The specific deletion of alloreactive T cells by mDC-FasL was confirmed by an unaffected proliferative response of surviving T cells towards allogeneic 'third-party' peripheral blood mononuclear cells in a third stimulation, or upon unspecific stimulation with anti-CD3/CD28 beads. The results of this study demonstrate that allospecifically activated T cells are efficiently eliminated by mDC-FasL, supporting further investigations to apply FasL-expressing 'killer-DC' as a novel strategy for the treatment of allograft rejection.
Collapse
Affiliation(s)
- Christian Schütz
- Department of Internal Medicine 1, University Medical Centre Regensburg, Regensburg, Germany.
| | | | | | | | | | | |
Collapse
|
6
|
Abstract
Immune regulation plays a critical role in controlling potentially dangerous inflammation and maintaining health. The Fas ligand/Fas receptor axis has been studied extensively as a mechanism of killing T cells and other cells during infections, autoimmunity, and cancer. FasL expression has been primarily attributed to activated T cells and NK cells. Evidence has emerged that B lymphocytes can express FasL and other death-inducing ligands, and can mediate cell death under many circumstances. Among B cell subsets, the expression of both Fas ligand and IL-10 is highest on the CD5(+) B cell population, suggesting that CD5(+) B cells may have a specialized regulatory function. The relevance of killer B cells to normal immune regulation, disease pathogenesis, and inflammation is discussed.
Collapse
Affiliation(s)
- Steven K Lundy
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA.
| |
Collapse
|
7
|
Chen J, Wang J, Li J, Wu Q, Lim FC, Yang P, Hsu HC, Curiel DT, Mountz JD. Enhancement of cytotoxic T-lymphocyte response in aged mice by a novel treatment with recombinant AdIL-12 and wild-type adenovirus in rapid succession. Mol Ther 2008; 16:1500-6. [PMID: 18545221 PMCID: PMC2575091 DOI: 10.1038/mt.2008.121] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
A decrease in the expression of Th1 cytokines has been associated with age-related decrease in cytotoxic T-lymphocyte (CTL) function. We utilized an E1-deleted adenovirus (Ad) vector to deliver the murine interleukin-12 (IL-12) gene in order to enhance the antivirus CTL response. Wild-type (WT) Ad was administered 3 days after AdIL-12 treatment, when IL-12 production was at its peak and the anti-Ad antibody response had not yet begun to develop. Before receiving AdIL-12 treatment, aged (18 month old) mice exhibited a 58% decrease in the number of virus-specific CTLs, and a 30% decrease in in vivo CTL activity as compared to young (2 month old) mice. After AdIL-12 treatment, aged mice displayed a greater increase in IL-12 expression and endogenous production of interferon-gamma than observed in young mice. When infected with WT Ad, these AdIL-12-treated aged mice exhibited an increased in vivo CTL response and an in vitro proliferative response that was similar to those in young mice. The frequencies of occurrence of D(b)-E1Bp(+)CD8(+) T cells in the spleen, liver, and lung in aged mice were higher than the corresponding values in young mice. These results indicate that IL-12 treatment significantly promotes the virus-specific CTL response in aged mice and, more importantly, specifically targets the virally infected organs, such as the liver and lung, promoting enhanced CTL activity against the virus.
Collapse
Affiliation(s)
- Jian Chen
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - John Wang
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jun Li
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Qi Wu
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Fei Chu Lim
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - PingAr Yang
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Hui-Chen Hsu
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - David T. Curiel
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Division of Human Gene Therapy, Gene Therapy Center; University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - John D. Mountz
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Veteran Administration Medical Center, Birmingham, Alabama, USA
| |
Collapse
|
8
|
Abstract
Several cell-based immunotherapy strategies have been developed to specifically modulate T cell-mediated immune responses. These methods frequently rely on the utilization of tolerogenic cell-based antigen-presenting cells (APCs). However, APCs are highly sensitive to cytotoxic T-cell responses, thus limiting their therapeutic capacity. Here, we describe a novel bead-based approach to modulate T-cell responses in an antigen-specific fashion. We have generated killer artificial APCs (kappaaAPCs) by coupling an apoptosis-inducing alpha-Fas (CD95) IgM mAb together with HLA-A2 Ig molecules onto beads. These kappaaAPCs deplete targeted antigen-specific T cells in a Fas/Fas ligand (FasL)-dependent fashion. T-cell depletion in cocultures is rapidly initiated (30 minutes), dependent on the amount of kappaaAPCs and independent of activation-induced cell death (AICD). kappaaAPCs represent a novel technology that can control T cell-mediated immune responses, and therefore has potential for use in treatment of autoimmune diseases and allograft rejection.
Collapse
|
9
|
Kim SH, Bianco NR, Shufesky WJ, Morelli AE, Robbins PD. Effective Treatment of Inflammatory Disease Models with Exosomes Derived from Dendritic Cells Genetically Modified to Express IL-4. THE JOURNAL OF IMMUNOLOGY 2007; 179:2242-9. [PMID: 17675485 DOI: 10.4049/jimmunol.179.4.2242] [Citation(s) in RCA: 145] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In this study, we demonstrate that genetically modified bone marrow-derived dendritic cells (DC) and exosomes derived from the DC, expressing either secreted IL-4 or membrane-bound IL-4, can reduce the severity and the incidence of established collagen-induced arthritis and inhibit inflammation of delayed-type hypersensitivity (DTH) in mice. The ability of the DC and DC-derived exosomes to suppress the DTH response was MHC class II and, in part, Fas ligand/Fas dependent. The DC-derived exosomes were internalized by CD11c(+) DC in the dermis at the site of injection and in the draining lymph node as well as by CD11c(+) DC and F4/80(+) macrophages in the spleen. Moreover, adoptive transfer of CD11c(+) or CD3(+) splenic cells from mice treated with exosomes showed significant reduction of footpad swelling in the DTH model. These results demonstrate that administration of DC/IL-4 or exosomes derived from DC/IL-4 are able to modulate the activity of APC and T cells in vivo through a MHC class II and partly Fas ligand/Fas-dependent mechanism, resulting in effective treatment of established collagen-induced arthritis and suppression of the DTH inflammatory response. Thus, APC-derived exosomes could be used therapeutically for the treatment of autoimmune disease and inflammatory disorders.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigens, CD1/genetics
- Antigens, CD1/immunology
- Arthritis, Experimental/genetics
- Arthritis, Experimental/immunology
- Arthritis, Experimental/therapy
- Autoimmune Diseases/genetics
- Autoimmune Diseases/immunology
- Autoimmune Diseases/therapy
- Bone Marrow Cells/immunology
- CD3 Complex/immunology
- Dendritic Cells/immunology
- Dermis/immunology
- Gene Expression
- Histocompatibility Antigens Class II/genetics
- Histocompatibility Antigens Class II/immunology
- Hypersensitivity, Delayed/genetics
- Hypersensitivity, Delayed/immunology
- Hypersensitivity, Delayed/therapy
- Inflammation/genetics
- Inflammation/immunology
- Inflammation/therapy
- Interleukin-4/genetics
- Interleukin-4/immunology
- Mice
- Mice, Inbred DBA
- Spleen/immunology
- T-Lymphocytes/immunology
- Transduction, Genetic
Collapse
Affiliation(s)
- Seon Hee Kim
- Department of Molecular Genetics and Biochemistry, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261
| | | | | | | | | |
Collapse
|
10
|
Pearl-Yafe M, Yolcu ES, Stein J, Kaplan O, Yaniv I, Shirwan H, Askenasy N. Fas ligand enhances hematopoietic cell engraftment through abrogation of alloimmune responses and nonimmunogenic interactions. Stem Cells 2007; 25:1448-55. [PMID: 17363551 DOI: 10.1634/stemcells.2007-0013] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Early after transplantation, donor lineage-negative bone marrow cells (lin(-) BMC) constitutively upregulated their expression of Fas ligand (FasL), suggesting an involvement of the Fas/FasL axis in engraftment. Following the observation of impaired engraftment in the presence of a dysfunctional Fas/FasL axis in FasL-defective (gld) donors or Fas-defective (lpr) recipients, we expressed a noncleavable FasL chimeric protein on the surface of donor lin(-) BMC. Despite a short life span of the protein in vivo, expression of FasL on the surface of all the donor lin(-) BMC improved the efficiency of engraftment twofold. The FasL-coated donor cells efficiently blunted the host alloimmune responses in primary recipients and retained their hematopoietic reconstituting potential in secondary transplants. Surprisingly, FasL protein improved the efficiency of engraftment in syngeneic transplants. The deficient engraftment in lpr recipients was not reversed in chimeric mice with Fas(-) stroma and Fas(+) BMC, demonstrating that the host marrow stroma was also a target of donor cell FasL. Hematopoietic stem and progenitor cells are insensitive to Fas-mediated apoptosis and thus can exploit the constitutive expression of FasL to exert potent veto activities in the early stages of engraftment. Manipulation of the donor cells using ectopic FasL protein accentuated the immunogenic and nonimmunogenic interactions between the donor cells and the host, alleviating the requirement for a megadose of transplanted cells to achieve a potent veto effect. Disclosure of potential conflicts of interest is found at the end of this article.
Collapse
Affiliation(s)
- Michal Pearl-Yafe
- Frankel Laboratory, Center for Stem Cell Research, Schneider Children's Medical Center of Israel, 14 Kaplan Street, Petach Tikva, Israel
| | | | | | | | | | | | | |
Collapse
|
11
|
Chen J, Hsu HC, Zajac AJ, Wu Q, Yang P, Xu X, McPherson SA, Li J, Curiel DT, Mountz JD. In vivo analysis of adenovirus-specific cytotoxic T lymphocyte response in mice deficient in CD28, fas ligand, and perforin. Hum Gene Ther 2006; 17:669-82. [PMID: 16776575 DOI: 10.1089/hum.2006.17.669] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Adenoviruses (Ad) have been extensively studied as gene delivery vectors in gene therapy and as vaccine carriers. The cell-mediated cytotoxicity induced by Ad is of great interest in both applications. However, the mechanism underlying Ad-specific cytotoxic T lymphocyte (CTL) generation and effector function remains unclear. In this study, we used a novel MHC class I tetramer and an in vivo CTL assay to examine the role of CD28, perforin, Fas ligand (FasL), and TNF-alpha in the generation and function of Ad-specific CTLs in vivo. During the primary response, there was a significant defect in both the generation and in vivo effector function of Ad-specific CTLs in CD28-/- mice, but not in CD4+ T cell-depleted mice or CD4-/- mice. The relative role of CTL effector molecules was assayed by in vivo CTL assay in perforin- or FasL-mutant mice, using donor cells from Fas-deficient or TNFR1/TNFR2-deficient mice. The results indicated that the in vivo CTL activity is mediated mainly by perforin. In the absence of perforin, production of FasL, but not TNF-alpha, by the CTLs results in lower level Ad-specific killing of target cells. These results provide important implications concerning the development of safe and effective Ad vectors for gene therapy and vaccines.
Collapse
Affiliation(s)
- Jian Chen
- Department of Medicine, University of Alabama at Birmingham, and Veterans Administration Medical Center, Birmingham, AL 35294, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Chen J, Hsu HC, Zajac AJ, Wu Q, Yang P, Xu X, McPherson SA, Li J, Curiel DT, Mountz JD. In Vivo Analysis of Adenovirus-Specific Cytotoxic T Lymphocyte Response in Mice Deficient in CD28, Fas Ligand, and Perforin. Hum Gene Ther 2006. [DOI: 10.1089/hum.2006.17.ft-217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
13
|
Chuang YH, Suen JL, Chiang BL. Fas-ligand-expressing adenovirus-transfected dendritic cells decrease allergen-specific T cells and airway inflammation in a murine model of asthma. J Mol Med (Berl) 2006; 84:595-603. [PMID: 16565865 DOI: 10.1007/s00109-006-0047-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2005] [Revised: 11/08/2005] [Accepted: 01/10/2006] [Indexed: 01/19/2023]
Abstract
T cells expressing a type-2 T helper profile of cytokines (Th2 cells) have been demonstrated to play an important role in the initiation and progression of allergic asthma, and it is well known that Fas ligand (FasL) induces apoptosis when bound to its receptor, Fas. In the present study, we examined the possibility of modulating asthma manifestations by dendritic cells (DCs) genetically engineered to express FasL (DC-FasL), which could deliver a death signal to T cells in an antigen-specific manner. The delivery of DC-FasL into ovalbumin (OVA)-immunized allergic mice decreased the airway hyper-responsiveness (AHR). Moreover, we established a mouse model of airway inflammation by using an adoptive transfer of Th2 cells derived from ovalbumin T cell receptor transgenic mice to study the effect of DC-FasL on airway reactivity. The administration of DC-FasL in Th2-cell-induced allergic mice had significantly decreased AHR, airway inflammation, and IL-4, IL-5 and IL-13 production. Furthermore, the numbers of OVA-specific T cells were decreased in the lung of mice receiving DC-FasL. These results demonstrate that FasL-expressing dendritic cells might be applied for the modulation of allergic responses.
Collapse
Affiliation(s)
- Ya-Hui Chuang
- Graduate Institute of Immunology, National Taiwan University, Taiwan, Republic of China
| | | | | |
Collapse
|
14
|
Kim SH, Bianco N, Menon R, Lechman ER, Shufesky WJ, Morelli AE, Robbins PD. Exosomes Derived from Genetically Modified DC Expressing FasL Are Anti-inflammatory and Immunosuppressive. Mol Ther 2006; 13:289-300. [PMID: 16275099 DOI: 10.1016/j.ymthe.2005.09.015] [Citation(s) in RCA: 169] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2005] [Revised: 09/01/2005] [Accepted: 09/06/2005] [Indexed: 12/30/2022] Open
Abstract
We previously have demonstrated the ability of primary murine bone marrow-derived DC (BM-DC), genetically modified by adenoviral infection to express FasL, to inhibit progression of established collagen-induced arthritis (CIA) following systemic delivery. Here we demonstrate that exosomes derived from genetically modified BM-DC expressing FasL are able to inhibit inflammation in a murine footpad model of delayed-type hypersensitivity (DTH). Local administration of exosomes derived from DC expressing FasL (Exo/FasL) as well as the parental DC/FasL resulted in a significant reduction in swelling in both the treated and the untreated distal paw. However, both the DC/FasL and the Exo/FasL were unable to suppress the DTH response in lpr (Fas-deficient) mice. Gene transfer of FasL to BM-DC from gld (FasL-deficient) mice resulted in restoration of the ability of DC as well as DC-derived exosomes to suppress DTH. The ability of DC-derived exosomes and DC to suppress DTH responses was antigen specific and MHC class II dependent, but class I independent. The injected exosomes were found to be internalized into CD11c(+) cells at the site of injection and in the draining popliteal lymph node. Systemic injection of exosome/FasL into mice with established CIA resulted in significant disease amelioration. These results demonstrate that both systemic and local administration of exosomes derived from FasL-expressing DC are able to suppress antigen-specific immune responses through an MHC class II-dependent pathway, resulting in effective and sustained treatment of established collagen-induced arthritis and suppression of the DTH inflammatory response. These results suggest that DC/FasL-derived exosomes could be used clinically for the treatment of inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Seon Hee Kim
- Department of Molecular Genetics and Biochemistry, University of Pittsburgh School of Medicine, PA 15261, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Chen J, Zajac AJ, McPherson SA, Hsu HC, Yang P, Wu Q, Xu X, Wang X, Fujihashi K, Curiel DT, Mountz JD. Primary adenovirus-specific cytotoxic T lymphocyte response occurs after viral clearance and liver enzyme elevation. Gene Ther 2005; 12:1079-88. [PMID: 15815704 DOI: 10.1038/sj.gt.3302494] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The virus-specific cytotoxic T lymphocyte (CTL) response is a major obstacle to effective delivery of adenovirus gene therapy. However, its relative role in viral clearance, transgene elimination and hepatotoxicity remains unclear. In this paper, we present an analysis of viral clearance and liver toxicity in relation to the induction of the virus-specific CD8 T-cell response revealed by an MHC class I tetramer. A surprisingly high number of tetramer+ CD8 T cells were found in the liver and lung and reached peak values at days 8 and 10, respectively, post-infection. Nearly 100% of these tetramer+ CD8 T cells expressed high levels of granzyme B and IFNgamma. Remarkably, liver viral load and liver enzyme elevation peaked early, at days 2 and 4, respectively, post-infection, before the specific CTL response was detectable. After generation of CTLs, there was only minimal liver damage or further decrease in virus titer. These results indicated that the primary peak response of tetramer+ CTLs does not correlate with the elimination of adenovirus or liver cytotoxic response.
Collapse
Affiliation(s)
- J Chen
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294-0007, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Mizutori Y, Saitoh O, Eguchi K, Nagayama Y. Adenovirus encoding the thyrotropin receptor A-subunit improves the efficacy of dendritic cell-induced Graves' hyperthyroidism in mice. J Autoimmun 2005; 26:32-6. [PMID: 16242303 DOI: 10.1016/j.jaut.2005.08.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2005] [Revised: 06/26/2005] [Accepted: 08/24/2005] [Indexed: 01/22/2023]
Abstract
Stimulating the immune system by in vivo expression of the thyrotropin receptor (TSHR) is an efficient means to induce Graves' disease experimentally. For example, BALB/c mice injected with dendritic cells (DCs) infected with adenovirus encoding the full-length TSHR (AdTSHR) develop hyperthyroidism, albeit at a low incidence (36%). Recent observations suggest that the shed TSHR A-subunit, rather than the full-length receptor, is the autoantigen responsible for initiating/enhancing immune responses leading to thyroid stimulating antibodies (TSAb) and hyperthyroidism. Therefore, we attempted to improve the efficacy of the DC-based approach for Graves' disease using adenovirus encoding the TSHR A-subunit (AdTSHR289). Three injections of DCs infected with AdTSHR289 induced hyperthyroidism in 70% of BALB/c mice, approximately twice the disease induction rate with AdTSHR. TSAb activity was detected in most hyperthyroid mice, whereas virtually all immunized mice developed antibodies that inhibit [125I]TSH binding to the TSHR or recognize linear or conformational epitopes on the TSHR. TSHR antibodies were of IgG1 and IgG2a, indicating mixed T-helper type 1 (Th1)/Th2 immune responses. In conclusion, immunization with DC infected with adenovirus expressing the TSHR A-subunit is a highly efficient protocol to induce Graves' hyperthyroidism in BALB/c mice. This improved model will permit studies of the pathogenic role and therapeutic potential of DCs in Graves' hyperthyroidism.
Collapse
Affiliation(s)
- Yumiko Mizutori
- Department of Medical Gene Technology, Atomic Bomb Disease Institute, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | | | | | | |
Collapse
|
17
|
Askenasy N, Yolcu ES, Yaniv I, Shirwan H. Induction of tolerance using Fas ligand: a double-edged immunomodulator. Blood 2005; 105:1396-404. [PMID: 15486063 DOI: 10.1182/blood-2004-06-2364] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
AbstractApoptosis mediated by Fas ligand (FasL) interaction with Fas receptor plays a pivotal regulatory role in immune homeostasis, immune privilege, and self-tolerance. FasL, therefore, has been extensively exploited as an immunomodulatory agent to induce tolerance to both autoimmune and foreign antigens with conflicting results. Difficulties associated with the use of FasL as a tolerogenic factor may arise from (1) its complex posttranslational regulation, (2) the opposing functions of different forms of FasL, (3) different modes of expression, systemic versus localized and transient versus continuous, (4) the level and duration of expression, (5) the sensitivity of target tissues to Fas/FasL-mediated apoptosis and the efficiency of antigen presentation in these tissues, and (6) the types and levels of cytokines, chemokines, and metalloproteinases in the extracellular milieu of the target tissues. Thus, the effective use of FasL as an immunomodulator to achieve durable antigen-specific immune tolerance requires careful consideration of all of these parameters and the design of treatment regimens that maximize tolerogenic efficacy, while minimizing the non-tolerogenic and toxic functions of this molecule. This review summarizes the current status of FasL as a tolerogenic agent, problems associated with its use as an immunomodulator, and new strategies to improve its therapeutic potential.
Collapse
Affiliation(s)
- Nadir Askenasy
- Frankel Laboratory, Center for Stem Cell Research, Department of Pediatric Hematology-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel 49202.
| | | | | | | |
Collapse
|
18
|
Xu X, Zhang HG, Liu ZY, Wu Q, Yang PA, Sun SH, Chen J, Hsu HC, Mountz JD. Defective Clearance of Adenovirus in IRF-1-/- Mice Associated with Defects in NK and T Cells but not Macrophages. Scand J Immunol 2004; 60:89-99. [PMID: 15238077 DOI: 10.1111/j.0300-9475.2004.01461.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A replication-defective adenovirus-LacZ recombinant virus (AdLacZ) was injected intravenously into IRF-1(-/-) mice and wild-type mice to characterize the contribution of IRF-1 to the immune-mediated clearance of Ad vector. Compared with wild-type mice, IRF-1(-/-) mice expressed higher levels of the LacZ gene product in the liver. After infusion of the AdLacZ, the expression of IRF-1 mRNA was upregulated in the liver of wild-type mice, but not in IRF-1(-/-) mice. Both spleen and liver mononuclear cells from IRF-1(-/-) mice initially exhibited a markedly lower number of NK, NK-T and CD8 T cells. At day 7 after the administration of AdLacZ, there was a significantly increased population of NK, NK-T and CD8 T cells in both spleen and liver, and also CD11b(+) cells in liver of IRF-1(-/-) mice, compared with the increased in wild-type mice. As IRF-1 is an important signal for production of IFN-gamma by CD8 T and NK cells as well as production of IL-12 by CD11b(+) cells, we determined whether there were lower levels of these cytokines in IRF-1(-/-) mice after Ad challenge. Surprisingly, there were lower levels of IL-12, but higher levels of IFN-gamma and IL-18 in IRF-1(-/-) compared with wild-type mice at day 7 after administration with AdLacZ. These results indicate that delayed clearance of Ad is associated with partial correction of defects of the NK, NK-T and CD8 T cells and increased production of IFN-gamma and IL-18 in IRF-1(-/-) mice.
Collapse
Affiliation(s)
- X Xu
- Department of Medicine, The University of Alabama at Birmingham, 701 S. 19th Street, Birmingham, AL 35294-0007, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Sun Z, Wada T, Hoshino S, Uchikura K, Klein AS. Immunomodulatory Role of Kupffer Cell in Liver Allografts. COMPARATIVE HEPATOLOGY 2004; 3 Suppl 1:S32. [PMID: 14960184 PMCID: PMC2410251 DOI: 10.1186/1476-5926-2-s1-s32] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Zhaoli Sun
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tatehiko Wada
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sumito Hoshino
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Keiichiro Uchikura
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Andrew S Klein
- Department of Surgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
20
|
Hoves S, Krause SW, Herfarth H, Halbritter D, Zhang HG, Mountz JD, Schölmerich J, Fleck M. Elimination of activated but not resting primary human CD4 and CD8 T cells by Fas ligand (FasL/CD95L)-expressing Killer-dendritic cells. Immunobiology 2004; 208:463-75. [PMID: 15124861 DOI: 10.1078/0171-2985-00293] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Dendritic cells (DC) genetically engineered to express high levels of Fas ligand (FasL/CD95L) have been demonstrated to delete T cells in an antigen specific manner in several different animal models in vivo. However, the immunomodulatory capacity of primary human FasL-expressing Killer-DC has not been determined. Therefore, human Killer-DC were generated from mature monocyte-derived DC using the inducible CRE/LoxP adenoviral vector system, and the immunoregulatory capacity of these cells was analyzed in cocultures with primary human T cells in vitro. Combined transductions of DC by AdloxPFasL and AxCANCre resulted in FasL expression in > 70% of DC without affecting the mature phenotype. Proliferation of activated primary human T cells was inhibited up to 80% in cocultures with FasL-expressing DC but not EGFP-transduced DC, which was due to induction of apoptosis in activated but not resting CD4+ and CD8+ T cells. Apoptosis induced by Killer-DC could be blocked by an anti-FasL-antibody in a dose dependent fashion. The present results demonstrate that FasL-expressing Killer-DC eliminate activated but not resting primary human CD4+ and CD8+ T cells by induction of Fas-mediated apoptosis supporting the concept to apply Killer-DC as a novel strategy for the treatment of T cell-dependent autoimmune disease and allograft rejection in humans.
Collapse
Affiliation(s)
- Sabine Hoves
- The University of Regensburg, Department of Internal Medicine I, 93042 Regensburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Zhang HG, Hsu HC, Yang PA, Yang X, Wu Q, Liu Z, Yi N, Mountz JD. Identification of multiple genetic loci that regulate adenovirus gene therapy. Gene Ther 2003; 11:4-14. [PMID: 14681692 DOI: 10.1038/sj.gt.3302136] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
A key aspect of the immune response to adenovirus (Ad) gene therapy is the generation of a cytotoxic T-cell (CTL) response. To better understand the genetic network underlying these events, 20 strains of C57BL/6 x DBA/2 (BXD) recombinant inbred (RI) mice were administered with AdLacZ and analyzed at days 7, 21, 30, and 50 for liver beta-galactosidase (LacZ) expression and CTL response. Sera levels of interferon gamma (IFN-gamma), tumor necrosis factor-alpha (TNF-alpha), and interleukin-6 (IL-6) were analyzed at different times after AdLacZ. There was a distinct strain-dependent expression of LacZ, which was strongly correlated with the CTL response. Among the five BXD RI strains that exhibited significantly prolonged LacZ expression, four also exhibited a marked defect in the production of Ad-specific CTL. There was a strong correlation between the sera levels of IFN-gamma, TNF-alpha, and IL-6, but cytokine responses were not significantly correlated with LacZ expression or the CTL response. Quantitative trait loci regulating LacZ on day 30 were found on chromosome (Chr) 19 (33 cM) and Chr 15 (42.8 cM). Cytotoxicity mapped to Chr 7 (41.0 and 57.4-65.2 cM), Chr 15 (61.7 cM), and Chr X (27.8 cM). IFN-gamma production mapped to Chr 18 (22, 27, and 32 cM) and Chr 11 (64.0 cM). TNF-alpha and IL-6 production mapped to Chr 6 (91.5 cM) Chr 9 (42.0 cM) and Chr 8 (52 and 73.0 cM). These results indicate that different strains of mice exhibit different pathways for effective clearance of AdLacZ depending on genetic polymorphisms and interactions at multiple genetic loci.
Collapse
Affiliation(s)
- H-G Zhang
- Department of Medicine, Division of Clinical Immunology and Rheumatology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Hoves S, Krause SW, Schölmerich J, Fleck M. The JAM-assay: optimized conditions to determine death-receptor-mediated apoptosis. Methods 2003; 31:127-34. [PMID: 12957570 DOI: 10.1016/s1046-2023(03)00122-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Apoptosis induced by interaction of members of the TNF-/TNF-receptor superfamily has been considered as a major mechanism of cell-mediated cytotoxicity. For functional analysis, the 51Cr release assay has been widely used, which requires loss of membrane integrity in the apoptotic target cell. However, loss of membrane integrity is a late event during apoptosis and therefore only late apoptotic cells will be detected by this method. In contrast, the JAM-assay first described by Polly Matzinger has been demonstrated to be more sensitive than the 51Cr release assay, since this method is dependent on DNA-fragmentation which precedes loss of membrane integrity in most apoptotic cells. The JAM-assay is easier to perform, less expansive, and safer than the current standard (51)Cr release assay. Therefore, this article will focus on optimized conditions of the JAM-assay to detect and quantitate Fas (CD95/Apo-1)-induced apoptosis as an example of death-receptor-mediated cytotoxicity.
Collapse
Affiliation(s)
- Sabine Hoves
- Department of Internal Medicine I, The University of Regensburg, 93042 Regensburg, Germany
| | | | | | | |
Collapse
|
23
|
Drachman DB, Wu JM, Miagkov A, Williams MA, Adams RN, Wu B. Specific Immunotherapy of Experimental Myasthenia by Genetically Engineered APCs. Ann N Y Acad Sci 2003; 998:520-32. [PMID: 14592923 DOI: 10.1196/annals.1254.068] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Although treatment of MG with general immunosuppressive agents is often effective, it has important drawbacks, including suppression of the immune system as a whole, with the risks of infection and neoplasia, and numerous other adverse side effects. Ideally, treatment of MG should eliminate the specific pathogenic autoimmune response to AChR, without otherwise suppressing the immune system or producing other adverse side effects. Although antibodies to AChR are directly responsible for the loss of AChRs at neuromuscular junctions in MG, the AChR antibody response is T cell-dependent, and immunotherapy directed at T cells can abrogate the autoantibody response, with resulting benefit. As in other autoimmune diseases, the T cell response in MG is highly heterogeneous. The design of specific immunotherapy must take this heterogeneity into account and target the entire repertoire of AChR-specific T cells. We describe our investigation of a novel strategy for specific immunotherapy of MG, involving gene transfer to convert antigen-presenting cells (APCs) to "guided missiles" that target AChR-specific T cells, and that induce apoptosis and elimination of those T cells. This strategy uses the ability of APCs from a given individual to present the entire spectrum of AChR epitopes unique for that individual, and thereby to target the entire repertoire of antigen-specific T cells of the same individual. Using viral vectors, we have genetically engineered the APCs to process and present the most important domain of the AChR molecule, and to express a "warhead" of Fas ligand (FasL) to eliminate the activated AChR-specific T cells with which they interact. Our results show that the APCs express the appropriate gene products, and effectively and specifically eliminate AChR-specific T cells by the Fas/FasL pathway, while sparing T cells of other specificities.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/immunology
- Cell Death
- Cell Line
- Dendritic Cells
- Genetic Engineering/methods
- Humans
- Immunotherapy
- Lymph Nodes/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Myasthenia Gravis, Autoimmune, Experimental/chemically induced
- Myasthenia Gravis, Autoimmune, Experimental/therapy
- Myasthenia Gravis, Autoimmune, Experimental/veterinary
- Proteins/metabolism
- Rats
- Rats, Inbred Lew
- Receptors, Cholinergic/chemistry
- Receptors, Cholinergic/immunology
- Receptors, Cholinergic/metabolism
- Signal Transduction
- Spleen/cytology
- Spleen/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Time Factors
- fas Receptor/metabolism
Collapse
Affiliation(s)
- D B Drachman
- Neuromuscular Laboratory, Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland 21287-7519, USA.
| | | | | | | | | | | |
Collapse
|
24
|
Hoves S, Krause SW, Halbritter D, Zhang HG, Mountz JD, Schölmerich J, Fleck M. Mature but not immature Fas ligand (CD95L)-transduced human monocyte-derived dendritic cells are protected from Fas-mediated apoptosis and can be used as killer APC. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:5406-13. [PMID: 12759415 DOI: 10.4049/jimmunol.170.11.5406] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Several in vitro and animal studies have been performed to modulate the interaction of APCs and T cells by Fas (CD95/Apo-1) signaling to delete activated T cells in an Ag-specific manner. However, due to the difficulties in vector generation and low transduction frequencies, similar studies with primary human APC are still lacking. To evaluate whether Fas ligand (FasL/CD95L) expressing killer APC could be generated from primary human APC, monocyte-derived dendritic cells (DC) were transduced using the inducible Cre/Loxp adenovirus vector system. Combined transduction of DC by AdLoxpFasL and AxCANCre, but not single transduction with these vectors, resulted in dose- and time-dependent expression of FasL in >70% of mature DC (mDC), whereas <20% of immature DC (iDC) expressed FasL. In addition, transduction by AdLoxpFasL and AxCANCre induced apoptosis in >80% of iDC, whereas FasL-expressing mDC were protected from FasL/Fas (CD95/Apo-1)-mediated apoptosis despite coexpression of Fas. FasL-expressing mDC eliminated Fas(+) Jurkat T cells as well as activated primary T cells by apoptosis, whereas nonactivated primary T cells were not deleted. Induction of apoptosis in Fas(+) target cells required expression of FasL in DC and cell-to-cell contact between effector and target cell, and was not dependent on soluble FasL. Induction of apoptosis in Fas(+) target cells required expression of FasL in DC, cell-to-cell contact between effector and target cell, and was not dependent on soluble FasL. The present results demonstrate that FasL-expressing killer APC can be generated from human monocyte-derived mDC using adenoviral gene transfer. Our results support the strategy to use killer APCs as immunomodulatory cells for the treatment of autoimmune disease and allograft rejection.
Collapse
Affiliation(s)
- Sabine Hoves
- Department of Internal Medicine I, Regensburg, Germany
| | | | | | | | | | | | | |
Collapse
|
25
|
Sun Z, Wada T, Maemura K, Uchikura K, Hoshino S, Diehl AM, Klein AS. Hepatic allograft-derived Kupffer cells regulate T cell response in rats. Liver Transpl 2003; 9:489-97. [PMID: 12740792 DOI: 10.1053/jlts.2003.50091] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In liver transplantation, the development of tolerance is associated with an increased rate of apoptosis of T lymphocytes in the portal inflammatory infiltrate and the presence of an intragraft Th2-like T cell population. Underlying mechanisms are poorly understood. Kupffer cells (KC), which reside in the hepatic sinosoids, can directly interact with circulating T lymphocytes and thus are uniquely positioned to play a role in immunomodulation. In this study, the immunoregulatory effects of KC were investigated. We show that KC can significantly suppress T cell proliferation in mixed leukocyte reaction (MLR). Furthermore, KC express functional Fas ligand (FasL) and can induce apoptosis of Fas+ cells. This process can be blocked by addition of neutralizing anti-FasL antibody. Moreover, using an allogeneic liver transplant model we have determined that 1. KC recovered from chronically accepted hepatic allografts have increased FasL messenger RNA (mRNA) and protein expression and a greater ability to induce apoptosis of alloreactive T cells compared with KC recovered from an acute rejection model; 2. KC not only induce apoptosis of T cells, but also regulate cytokine production and Th2/Th3-like cytokine (interleukin [IL]-10 / transforming growth factor [TGF]-beta) mRNA expression in allogeneic MLR in vitro; and 3. administration of KC derived from chronically accepted liver allografts significantly prolongs the survival of hepatic allografts in an acute rejection model in an alloantigen-specific manner. In conclusion, these data implicate the possible role of KC-mediated regulation of T cell response in the induction of immune tolerance in liver allografts.
Collapse
Affiliation(s)
- Zhaoli Sun
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
The proteins of the tumor necrosis factor (TNF) receptor superfamily are a group of cell-surface receptors critically involved in the maintenance of homeostasis of the immune system. By interacting with their corresponding ligands, these receptors either induce cell death or promote cell survival of immune cells. The number of recognized members of the TNF receptor and ligand superfamily has expanded substantially in the last several years. More important, the biologic function of this group of proteins has been closely associated with the regulation of the immune response and the pathogenesis of autoimmune disease. Thus, the direct targeting of these receptors by either inducing apoptosis or blocking survival of autoimmune T and B cells may be an important therapeutic strategy in the treatment of autoimmune disease. This review summarizes the recent progress in immunobiology of the TNF receptor superfamily and focuses on our studies of three critical family members-FasL/Fas, TNF-related apoptosis-inducing ligand (TRAIL)/TRAIL-Rs, and B lymphocyte stimulator(BLyS)/BLyS-Rs--to demonstrate the therapeutic potential of targeting these receptors for the treatment of autoimmune disease.
Collapse
Affiliation(s)
- Tong Zhou
- Department of Medicine, University of Alabama at Birmingham, 35294, USA.
| | | | | |
Collapse
|
27
|
Zhan HG, Mountz JD, Fleck M, Zhou T, Hsu HC. Specific deletion of autoreactive T cells by adenovirus-transfected, Fas ligand-producing antigen-presenting cells. Immunol Res 2003; 26:235-46. [PMID: 12403361 DOI: 10.1385/ir:26:1-3:235] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Immune privilege is a unique strategy developed in several internal organs that can prevent the development of immune attack against these vital organs. One critical mechanism of immune privilege is utilization of Fas/FasL-mediated apoptosis to delete the invading T cells at the immune privilege sites. In this article, we describe the development and application of a unique cell-gene therapy to correct defective FasL-mediated apoptosis and autoimmune disease in autoimmune mice. This cell-gene therapy strategy using antigen-presenting cells (APCs) to express FasL is not only a therapeutic tool, but also has allowed us to understand the complexity of T cell regulation and the concept of eliminating T cells in the spleen, lymph node, or elsewhere in vivo to regulate the homeostasis of the peripheral T cell response. In this regard, the FasL-expressing APCs can be considered as circulating and regulatable immune privilege sites. Our studies provide substantial evidence that FasL-expressing APCs can be introduced exogenously without liver toxicity to eliminate infiltrating T cells and prevent the development of immune attack in lung, liver, kidney, joint, and salivary gland. Therefore, given the hazardous potential of persistent T cell invasion at the local inflammatory site, it is tempting to speculate that such an endogenous control mechanism occurs normally in vivo to limit a chronic T cell inflammatory response.
Collapse
Affiliation(s)
- Huang-Ge Zhan
- Department of Medicine, University of Alabama at Birmingham, 35294, USA
| | | | | | | | | |
Collapse
|
28
|
Giannoukakis N, Robbins PD. Gene and cell therapies for diabetes mellitus: strategies and clinical potential. BioDrugs 2003; 16:149-73. [PMID: 12102644 DOI: 10.2165/00063030-200216030-00001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The last 5 years have witnessed an explosion in the use of genes and cells as biomedicines. While primarily aimed at cancer, gene engineering and cell therapy strategies have additionally been used for Mendelian, neurodegenerative and metabolic disorders. The main focus of gene and cell therapy strategies in metabolism has been diabetes mellitus. This disease is a disorder of glucose homeostasis, either due to the immune-mediated eradication of pancreatic beta cells in the islets of Langerhans (type 1 diabetes) or resulting from insulin resistance and obesity syndromes where the insulin-producing capability of the beta cell is ultimately exhausted in the face of insensitivity to the effects of insulin in the peripheral glucose-utilising tissues (type 2 diabetes). A significant number of animal studies have demonstrated the potential in restoring normoglycaemia by islet transplantation in the context of immunoregulation achieved by gene transfer of immunoregulatory genes to allo- and xenogeneic islets ex vivo. Additionally, gene and cell therapy has also been used to induce tolerance to auto- and alloantigens and to generate the tolerant state in autoimmune rodent animal models of type 1 diabetes or rodent recipients of allogeneic/xenogeneic islet transplants. The achievements of gene and cell therapy in type 2 diabetes are less evident, but seminal studies promise that this modality can be relevant to treat and perhaps prevent the underlying causes of the disease. Here we present an overview of the current status of gene and cell therapy for type 1 and 2 diabetes and we propose potential therapeutic options that could be clinically useful. For type 1 diabetes, transplantation of islets engineered to evade or suppress the recipient immune response is the most readily-available technology today. A number of gene delivery vectors encoding proteins that impair a variety of immune cells have already been examined and proven versatile. More challenging but, nonetheless, just over the horizon are attempts to promote tolerance to islet allografts. Type 2 diabetes will likely require a better understanding of the processes that determine insulin sensitivity in the periphery. Targeting tissues such as muscle and fat with vectors encoding genes whose products promote insulin sensitivity and glucose uptake is an approach that does not carry with it the side-effects often associated with pharmacologic agents currently in use. In the end, progress in vector design, elucidation of antigen-specific immunity and insulin sensitivity will provide the framework for gene drug use in the treatment of type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Nick Giannoukakis
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA.
| | | |
Collapse
|
29
|
Mountz JD. Adeno-associated viruses: Monkey see, monkey do. Gene Ther 2003. [DOI: 10.1038/sj.gt.3301922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
30
|
Abstract
Autoimmune disease in the elderly is hypothesized to be caused by an imbalance in T-cell expansion and deletion after an encounter with self-antigens. A decrease in thymic output leads to a decreased pool of naive T cells in the periphery and to increased oligoclonal expansion of T cells. This expansion may be caused by stimulation with autoantigens that drive high-affinity interactions with self-antigens. Accumulation of presenescent, apoptosis-resistant, and proinflammatory T cells results in the growth of these autoreactive T cells. A decreased T-cell activation response that occurs with age leads to several defects that diminish the immune response.
Collapse
Affiliation(s)
- Hui-Chen Hsu
- Division of Clinical Immunology and Rheumatology, Department of Medicine, University of Alabama at Birmingham, 701 South 19th Street, Lyons Harrison Research Building 473, Birmingham, AL 35294, USA
| | | |
Collapse
|
31
|
Chen A, Zheng G, Tykocinski ML. Quantitative interplay between activating and pro-apoptotic signals dictates T cell responses. Cell Immunol 2003; 221:128-37. [PMID: 12747954 DOI: 10.1016/s0008-8749(03)00069-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Antigen-presenting cells (APC) can express surface ligands with both T cell activating and inhibitory capacities, prompting the question of how responding T cells integrate opposing trans signals concurrently delivered by APC. To address this question in a quantitative fashion, we turned to protein transfer as a unique experimental approach that is well-suited for addressing such questions from a quantitative standpoint. Costimulatory (either B7-1*Fc(gamma1) or Fc(gamma1)*4-1BBL) and pro-apoptotic (Fc(gamma1)*FasL) Fc fusion proteins were quantitatively "painted" in varying ratios onto surrogate APC pre-coated with palmitated-protein A, the latter serving as a surface anchor. Evaluating the signaling potential of these various painted cells in a standard in vitro T cell proliferation assay, we demonstrated that at a given level of TCR triggering, the quantitative balance between costimulator (B7-1 or 4-1BBL) and FasL dictates the magnitude of the proliferative T cell response. Furthermore, when the costimulator density is kept constant, there is also a quantitative balance between TCR-directed and FasL signals. Interesting species-specific nai;ve versus memory T cell subset differences emerged with regard to susceptibility to Fas-mediated apoptosis and costimulator:FasL opposition. Taken together, these data demonstrate for the first time a quantitative interplay between activating and pro-apoptotic trans signals that dictates the magnitude of T cell responses.
Collapse
Affiliation(s)
- Aoshuang Chen
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, 6 Gates Building, 3400 Spruce Street, Philadelphia, PA 19104-4283, USA
| | | | | |
Collapse
|
32
|
Zuraff-Perryman LA, Duke RC, Setser E, Nelson DP, Bellgrau D. Murine heart transgenic Fas ligand expression protects against allograft rejection. Transplant Proc 2002; 34:3396-8. [PMID: 12493481 DOI: 10.1016/s0041-1345(02)03621-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
33
|
de Grey ADNJ. Bioremediation meets biomedicine: therapeutic translation of microbial catabolism to the lysosome. Trends Biotechnol 2002; 20:452-5. [PMID: 12413818 DOI: 10.1016/s0167-7799(02)02062-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Lysosomal degradation of damaged macromolecules is imperfect: many cell types accumulate lysosomal aggregates with age. Some such deposits are known, or are strongly suspected, to cause age-related disorders such as atherosclerosis and neurodegeration. It is possible that they also influence the rate of aging in general. Lysosomal degradation involves extensive cooperation between the participating enzymes: each generates a substrate for others until breakdown of the target material to recyclable units (such as amino acids) is complete. Hence, the age-related accumulation of lysosomal aggregates might be markedly retarded, or even reversed, by introducing just a few bacterial or fungal enzymes -'xenohydrolases' - that can degrade molecules that our natural machinery cannot. This article examines the feasibility and biomedical potential of such lysosomal enhancement as an approach to retarding or treating age-related physiological decline and disease.
Collapse
|
34
|
Effective Treatment of Established Mouse Collagen-Induced Arthritis by Systemic Administration of Dendritic Cells Genetically Modified to Express FasL. Mol Ther 2002. [DOI: 10.1006/mthe.2002.0712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
35
|
Abstract
Vaccines against a number of diseases, including HIV, Epstein Barr virus, malaria and several cancers, are believed to require the coinduction of multiple alphabeta CD8+ cytotoxic T-lymphocyte responses that are directed towards a number of different target antigens. The difficulties associated with making large recombinant vaccines that contain numerous antigens has led to the development of alphabeta CD8+ cytotoxic T-lymphocyte polyepitope or polytope vaccine approach, where multiple (usually 8-10 amino acids long) alphabeta CD8+ cytotoxic T-lymphocyte epitopes, derived from several antigens are conjoined into single artificial constructs. Such polytope constructs can be delivered using a number of different vaccine vector modalities with each epitope in the construct emerging as individually immunogenic.
Collapse
Affiliation(s)
- Andreas Suhrbier
- Cooperative Research Center for Vaccine Technology, Queensland Institute of Medical Research, University of Queensland, Australia.
| |
Collapse
|
36
|
Olive M, Eisenlohr L, Flomenberg N, Hsu S, Flomenberg P. The adenovirus capsid protein hexon contains a highly conserved human CD4+ T-cell epitope. Hum Gene Ther 2002; 13:1167-78. [PMID: 12133270 DOI: 10.1089/104303402320138952] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The immunogenicity of adenovirus vectors remains a major obstacle to their safe and efficacious use for gene therapy. In order to identify T-cell epitopes directly from adenoviruses, four viral protein sequences were screened for the well-characterized 9-mer HLA-A2 binding motif. Peripheral blood mononuclear cells (PBMC) from healthy adults were tested for responses to 17 selected viral peptides using a short-term interferon-gamma ELISPOT assay. Memory T-cell responses were identified to a single peptide derived from the major capsid protein hexon in 5 of 6 HLA-A2-positive donors. Unexpectedly, responses to this hexon peptide were also detected in 4 of 6 HLA-A2-negative donors, and responder cells were identified as CD4(+) T cells by immunomagnetic depletion experiments. A longer 15-mer peptide, H910-924, was identified as the optimal CD4(+) T-cell epitope. This hexon epitope induces strong proliferative T-cell responses that can be blocked by a monoclonal antibody against HLA-DR, and molecular HLA typing of donors suggests that the peptide response is restricted by multiple HLA-DR alleles. Additionally, quantitative analysis of responses to H910-924 and whole adenovirus reveals that the frequency of circulating CD4(+) T cells specific for this single hexon epitope (mean = 61 per 10(6) PBMC) represents up to one third of the total adenovirus-specific T-cell response. Finally, comparison of hexon sequences from over 20 different human adenovirus serotypes indicates that H910-924 is highly conserved. In most individuals, therefore, T-cell responses to this hexon epitope will be induced by all adenovirus vectors, including "gutted" vectors packaged with capsid proteins and vectors based on different serotypes.
Collapse
Affiliation(s)
- Melanie Olive
- Center of Human Virology and Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | | | | | | | | |
Collapse
|
37
|
Matsuki Y, Li L, Hsu HC, Yang PA, Zheng R, Edwards CK, Chaudry IH, Zhang HG, Mountz JD. Soluble Fas gene therapy protects against Fas-mediated apoptosis of hepatocytes but not the lethal effects of Fas-induced TNF-alpha production by Kupffer cells. Cell Death Differ 2002; 9:626-35. [PMID: 12032671 DOI: 10.1038/sj.cdd.4401016] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2001] [Revised: 11/06/2001] [Accepted: 12/06/2001] [Indexed: 02/01/2023] Open
Abstract
The elevation of soluble Fas (sFas) in the sera of patients with liver disease suggests a role for sFas in the disease process; whether it is protective or not is controversial. To determine the effects of sFas on Fas-induced liver apoptosis, we manipulated mice to produce sFas by transfecting them in vivo with different amounts of an adenovirus that produces mouse sFas driven by the CMV promoter (AdsFas). Fas-mediated apoptosis was induced by administration of anti-mouse Fas (Jo2; 10 microg/mouse) one week later. The administration of AdsFas (10(3), 10(7), or 10(9) pfu/mouse), which was associated with only minimal side-effects, resulted in a significant reduction in the liver transaminase levels and mortality of the mice on challenge with Jo2, as compared to control mice treated with AdLacZ. However, the protective effect of AdsFas was not complete. The possibility that Jo2-induction of TNF-alpha in the Kupffer cells of the liver contributes to the pathology was therefore tested. Although administration of soluble TNF receptor (sTNFRI) alone did not protect the mice from the lethal effects of Jo2, administration of sTNFRI (200 microg/mouse) after infection with AdsFas (10(9) pfu/mouse) resulted in 100% survival of the mice on challenge with Jo2. To confirm that the production of TNF-alpha by Kupffer cells produce the lethal effects of Jo2 that remained after treatment with AdsFas, these cells were selectively ablated by treatment of the mice with gadolinium chloride prior to challenge with Jo2. This treatment greatly reduced early mortality and hepatocellular damage as well as TNF-alpha production 6 h after injection of Jo2. These results indicate that: (1) AdsFas prevents Jo2-induced apoptosis of hepatocytes; (2) In addition to mediating Fas-mediated apoptosis of hepatocytes, Jo2 can separately induce TNF-alpha production by Kupffer cells resulting in early mortality, and (3) Optimal protection from Jo2-induced mortality can be achieved by protection of liver cells by pretreatment with both AdsFas and sTNFRI.
Collapse
Affiliation(s)
- Y Matsuki
- Division of Clinical Immunology and Rheumatology, Department of Medicine, The University of Alabama at Birmingham, 35294-0007, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Zhang HG, Xie J, Xu L, Yang P, Xu X, Sun S, Wang Y, Curiel DT, Hsu HC, Mountz JD. Hepatic DR5 induces apoptosis and limits adenovirus gene therapy product expression in the liver. J Virol 2002; 76:5692-700. [PMID: 11991997 PMCID: PMC137014 DOI: 10.1128/jvi.76.11.5692-5700.2002] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A major limitation of adenovirus (Ad) gene therapy product expression in the liver is subsequent elimination of the hepatocytes expressing the gene therapy product. This elimination is caused by both necrosis and apoptosis related to the innate and cell-mediated immune response to the Ad. Apoptosis of hepatocytes can be induced by the innate immune response by signaling through death domain receptors on hepatocytes including the tumor necrosis factor alpha (TNF-alpha) receptor (TNFR), Fas, and death domain receptors DR4 and DR5. We have previously shown that blocking signaling through TNFR enhances and prolongs gene therapy product expression in the liver. In the present study, we constructed an Ad that produces a soluble DR5-Fc (AdsDR5), which is capable of neutralizing TNF-related apoptosis-inducing ligand (TRAIL). AdsDR5 prevents TRAIL-mediated apoptosis of CD3-activated T cells and decreases hepatocyte apoptosis after AdCMVLacZ administration and enhances the level and duration of lacZ transgene expression in the liver. In addition to blocking TRAIL and directly inhibiting apoptosis, AdsDR5 decreases production of gamma interferon (IFN-gamma) and TNF-alpha and decreases NK cell activation, all of which limit Ad-mediated transgene expression in the liver. These results indicate that (i) AdsDR5 produces a DR5-Fc capable of neutralizing TRAIL, (ii) AdsDR5 can reduce activation of NK cells and reduce induction of IFN-gamma and TNF-alpha after Ad administration, and (iii) administration of AdsDR5 can enhance Ad gene therapy in the liver.
Collapse
MESH Headings
- Adenoviruses, Human
- Animals
- Antigens, CD/metabolism
- Antigens, CD/pharmacology
- Apoptosis
- Apoptosis Regulatory Proteins
- Aspartate Aminotransferases/metabolism
- Cells, Cultured
- Cytomegalovirus/genetics
- Gene Expression
- Genetic Therapy
- Genetic Vectors
- Hepatocytes/cytology
- Humans
- Interferon-gamma/biosynthesis
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Lac Operon
- Liver/metabolism
- Lymphocyte Activation
- Membrane Glycoproteins/metabolism
- Membrane Glycoproteins/pharmacology
- Mice
- Mice, Inbred BALB C
- Promoter Regions, Genetic
- Receptors, TNF-Related Apoptosis-Inducing Ligand
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor, Type I
- Spleen/cytology
- T-Lymphocytes/cytology
- T-Lymphocytes/metabolism
- TNF-Related Apoptosis-Inducing Ligand
- Transgenes
- Tumor Necrosis Factor-alpha/biosynthesis
- Tumor Necrosis Factor-alpha/metabolism
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- Huang-Ge Zhang
- Division of Clinical Immunology and Rheumatology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Zhang HG, Yang P, Xie J, Liu Z, Liu D, Xiu L, Zhou T, Wang Y, Hsu HC, Mountz JD. Depletion of collagen II-reactive T cells and blocking of B cell activation prevents collagen II-induced arthritis in DBA/1j mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:4164-72. [PMID: 11937577 DOI: 10.4049/jimmunol.168.8.4164] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Collagen II (CII)-induced arthritis in DBA/1j mice is mediated by both CII-reactive T cells and anti-CII Ab-producing B cells. To determine the relative role of these processes in the development of arthritis, we specifically eliminated CII-reactive T cells by treating the mice with CII-pulsed syngeneic macrophages that had been transfected with a binary adenovirus system. These macrophages express murine Fas ligand in a doxycycline-inducible manner with autocrine suicide inhibited by concomitant expression of p35. The mice were treated i.v. with four doses of CII-APC-AdFasLp35Tet or a single dose of AdCMVsTACI (5 x 10(9) PFU), or both simultaneously, beginning 2 wk after priming with CII in CFA. Treatment with CII-APC-AdFasLp35Tet alone or in combination with a single dose of AdCMVsTACI prevented the development of CII-induced arthritis and T cell infiltration in the joint. The elimination of T cells was specific in that a normal T cell response was observed on stimulation with OVA after treatment with CII-APC-AdFasLp35Tet. Treatment with AdCMVsTACI alone prevented production of detectable levels of circulating anti-CII autoantibodies and reduced the severity of arthritis but did not prevent its development. These results indicate that the CII-reactive T cells play a crucial role in the development of CII-induced arthritis and that the anti-CII Abs act to enhance the development of CII-induced arthritis.
Collapse
MESH Headings
- Adenoviridae/genetics
- Administration, Oral
- Animals
- Antigen-Presenting Cells/transplantation
- Apoptosis/genetics
- Apoptosis/immunology
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- Arthritis, Experimental/prevention & control
- Autoantibodies/biosynthesis
- Autocrine Communication/genetics
- Autocrine Communication/immunology
- B-Lymphocytes/immunology
- Cartilage, Articular/metabolism
- Cartilage, Articular/pathology
- Cell Migration Inhibition
- Collagen Type II/administration & dosage
- Collagen Type II/immunology
- Cytomegalovirus/genetics
- Down-Regulation/genetics
- Down-Regulation/immunology
- Drug Therapy, Combination
- Fas Ligand Protein
- Female
- Genetic Vectors/administration & dosage
- Genetic Vectors/therapeutic use
- Inhibitor of Apoptosis Proteins
- Lymphocyte Activation/genetics
- Lymphocyte Depletion/methods
- Macrophages, Peritoneal/transplantation
- Membrane Glycoproteins/biosynthesis
- Membrane Glycoproteins/genetics
- Membrane Proteins
- Mice
- Mice, Inbred DBA
- Receptors, Tumor Necrosis Factor/biosynthesis
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/physiology
- Solubility
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/pathology
- Tetracycline Resistance/genetics
- Trans-Activators/genetics
- Transmembrane Activator and CAML Interactor Protein
- Viral Proteins/genetics
Collapse
|
40
|
Kusuhara M, Matsue K, Edelbaum D, Loftus J, Takashima A, Matsue H. Killing of naive T cells by CD95L-transfected dendritic cells (DC): in vivo study using killer DC-DC hybrids and CD4(+) T cells from DO11.10 mice. Eur J Immunol 2002; 32:1035-43. [PMID: 11920570 DOI: 10.1002/1521-4141(200204)32:4<1035::aid-immu1035>3.0.co;2-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Dendritic cells (DC) play the dual task of initiating cellular immunity against potentially harmful foreign antigens (Ag), while maintaining immunological tolerance to self-Ag and environmental Ag. As an approach to induce Ag-specific suppression, we and others introduced CD95 ligand (L) cDNA into DC. The resulting "killer" DC delivered apoptotic signals, instead of activation signals, to primed CD4(+) T cells in vitro and induced Ag-specific immunosuppression in vivo. To study the impact of killer DC on naive T cells, the fate of Ag-reactive T cells and the extent of their depletion after killer DC treatment, we performed in vitro and in vivo reconstitution experiments using: (a) killer DC-DC hybrids created between CD95L-transduced XS106 DC clone (A/J origin) and splenic DC from BALB/c mice, (b) CD4(+) T cells isolated from DO11.10 transgenic mice (BALB/c background), and (c) OVA(323-339) peptide as relevant Ag. Ovalbumin (OVA)-pulsed killer DC-DC hybrids inhibited DO11.10 T cell activation triggered by conventional DC, instead of inducing their activation. Rapid apoptosis of T cells was observed after co-culture with OVA-pulsed killer DC-DC hybrids, but not with non-pulsed killer DC-DC hybrids or OVA-pulsed control DC-DC hybrids. For in vivo reconstitution, (BALB/cxA/J)F1 mice received subcutaneous administration of killer DC-DC hybrids, followed by intravenous inoculation of DO11.10 T cells. Killer DC-DC hybrids migrated preferentially to draining lymph nodes albeit with relatively low efficiency (0.5-1% recovery) and they induced significant, but incomplete (30-40%) killing of DO11.10 T cells in this location. These results document the abilities of CD95L-transduced DC to trigger apoptosis of naive T cells in an Ag-specific manner, to overrule T cell activation signals delivered by conventional DC, and to reduce local frequencies of Ag-reactive T cells in vivo. Our data also uncover two major limitations (relatively low homing efficiency and incomplete elimination of Ag-reactive T cells) that remain to be overcome for clinical application of CD95L-transduced DC strategy.
Collapse
Affiliation(s)
- Masahiro Kusuhara
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, USA
| | | | | | | | | | | |
Collapse
|
41
|
Hsu HC, Zhang HG, Song GG, Xie J, Liu D, Yang PA, Fleck M, Wintersberger W, Zhou T, Edwards CK, Mountz JD. Defective Fas ligand-mediated apoptosis predisposes to development of a chronic erosive arthritis subsequent to Mycoplasma pulmonis infection. ARTHRITIS AND RHEUMATISM 2001; 44:2146-59. [PMID: 11592380 DOI: 10.1002/1529-0131(200109)44:9<2146::aid-art368>3.0.co;2-o] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE To determine whether defective T cell apoptosis is associated with the development of a chronic arthritis subsequent to mycoplasma infection, and to determine whether deletion of T cells can prevent the development of this arthritis. METHODS B6 wild-type (B6-+/+), B6-lpr/lpr, and B6-gld/gld mice were infected with Mycoplasma pulmonis. The severity of lymphocytic infiltration and joint damage was evaluated, and the degree of recovery of viable mycoplasma from the spleen and joints was determined. Antigen-presenting cells derived from Fas mutant lpr mice (lpr-APC) were transfected ex vivo with an adenovirus (Ad) vector to yield lpr-APC expressing high levels of Fas ligand (lpr-APC-AdFasL), which in turn were transferred intraperitoneally into M pulmonis-infected B6-gld/gld mice. The development of arthritis subsequent to M pulmonis infection and the induction of apoptosis of cells within the synovial tissue and lymph nodes of lpr-APC-AdFasL-treated B6-gld/gld mice were determined. RESULTS Infection of B6-lpr/lpr and B6-gld/gld mice with M pulmonis resulted in an acute-phase inflammation of the synovium that later developed into a chronic erosive arthritis. Similar infection of B6-+/+ mice resulted only in an acute joint inflammatory response that resolved. Chronic arthritis in B6-gld/gld mice and B6-lpr/lpr was not due to persistent infection, since there were no differences in the rates of clearance of M pulmonis from the joints of B6-gld/gld or B6-lpr/lpr mice compared with B6-+/+ mice. Treatment of infected B6-gld/gld mice with lpr-APC-AdFasL resulted in a significantly decreased incidence of chronic arthritis that was associated with a decrease in lymph node T cells, but not with apoptosis of synovial T cells or fibroblasts. CONCLUSION Defective Fas/FasL-mediated apoptosis of T cells is an important factor that rendered arthritis-resistant B6 mice susceptible to the development of a chronic erosive arthritis subsequent to mycoplasma infection. In vivo lpr-APC-AdFasL cell-gene therapy is a safe and effective method for inhibiting the development of this arthritis.
Collapse
Affiliation(s)
- H C Hsu
- The University of Alabama at Birmingham, 35294-0007, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Zhang HG, Wang YM, Xie JF, Liang X, Hsu HC, Zhang X, Douglas J, Curiel DT, Mountz JD. Recombinant adenovirus expressing adeno-associated virus cap and rep proteins supports production of high-titer recombinant adeno-associated virus. Gene Ther 2001; 8:704-12. [PMID: 11406765 DOI: 10.1038/sj.gt.3301454] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2000] [Accepted: 02/07/2001] [Indexed: 11/09/2022]
Abstract
It has been difficult to produce a chimeric vector containing both Ad and AAV rep and cap, and to grow such chimeric vectors in 293 cells. By recombination in vitro in a bacterial host, we were able to produce recombinant plasmid AdAAV (pAdAAVrep-cap), which could be used to generate recombinant AdAAV (rAdAAVrep-cap) after transfection into 293 cells. A recombinant adenovirus, rAdAAVGFP, in which the green fluorescent protein (GFP) gene is flanked by the AAV terminal repeats cloned into the E1-deleted site of Ad was also generated. Co-infection of rAdAAVrep-cap together with rAdAAVGFP into 293 cells resulted in production of high titers of rAAV expressing GFP. It was noted that the titer of rAdAAVrep-cap was lower than the titer of control AdCMVLacZ. The lower titer of rAdAAvrep-cap was associated with expression of Rep protein. Non-homologous recombination occurs after high passage and results in deletions within the AAV rep genes. These results indicate that (1) rAdAAVrep-cap can be produced; (2) rAdAAVrep-cap + rAdAAVGFP is a convenient and efficient way to transfect 293 cells to grow high titer rAAV; and (3) frozen stock is required to avoid propagation of rep-deleted pAdAAVrep-cap.
Collapse
Affiliation(s)
- H G Zhang
- The University of Alabama at Birmingham, Department of Medicine, Division of Clinical Immunology and Rheumatology, Birmingham, AL 35294, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Wu B, Wu JM, Miagkov A, Adams RN, Levitsky HI, Drachman DB. Specific immunotherapy by genetically engineered APCs: the "guided missile" strategy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:4773-9. [PMID: 11254740 DOI: 10.4049/jimmunol.166.7.4773] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We tested the hypothesis that APCs genetically engineered to present an Ag and to express Fas ligand (FasL) simultaneously can target and eliminate Ag-specific T cells. Transgenic T cells specific for influenza hemagglutinin (HA) were used as targets. We prepared recombinant vaccinia virus vectors (VVV) to transfer the gene constructs individually or simultaneously into APCs. We prevented unwanted viral replication by attenuating the VVVs with psoralen-UV light treatment. For presentation of the HA Ag, APCs were transduced with cDNA for HA flanked by sequences of the lysosome-associated membrane protein that direct efficient processing and presentation of the Ag by APCs. As a "warhead" for the APCs, we transduced them with the gene for FasL, which induces apoptosis of Fas-expressing activated T cells. To protect the transduced APCs from self-destruction by FasL, we transferred cDNA for a truncated form of Fas-associated death domain, which inhibits Fas-mediated cell death. Our results show that the engineered APCs effectively expressed the genes of interest. APCs transduced with VVV carrying all three gene constructs specifically killed HA-transgenic T cells in culture. Coculture with T cells specific for an unrelated Ag (OVA) had no significant effect. Our in vitro findings show that APCs can be genetically engineered to target and kill Ag-specific T cells and represent a promising novel strategy for the specific treatment of autoimmune diseases.
Collapse
Affiliation(s)
- B Wu
- Neuromuscular Research Laboratory, Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | | | | | | | | | | |
Collapse
|
44
|
Fleck M, Zhang HG, Kern ER, Hsu HC, Müller-Ladner U, Mountz JD. Treatment of chronic sialadenitis in a murine model of Sjögren's syndrome by local fasL gene transfer. ARTHRITIS AND RHEUMATISM 2001; 44:964-73. [PMID: 11315936 DOI: 10.1002/1529-0131(200104)44:4<964::aid-anr154>3.0.co;2-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVE Infection of Fas (Fas/CD95)-mutant C57BL/6 (B6)-lpr/lpr mice with murine cytomegalovirus (MCMV) leads to a chronic sialadenitis similar to that of Sjögren's syndrome (SS). The aim of this study was to evaluate whether chronic sialadenitis would also occur in Fas ligand (FasL/CD95L)-mutant B6-gld/gld mice upon infection with MCMV and whether the expression of FasL by local gene transfer using recombinant adenoviral vectors would be an effective therapeutic strategy. METHODS B6-gld/gld mice were infected intraperitoneally with MCMV, and salivary glands were analyzed histologically at different time points. For treatment of sialadenitis, recombinant adenoviral vectors expressing the fasL gene (AdLoxpFasL + AxCANCre) or the lacZ gene (AdCMVLacZ) were locally injected into the salivary glands of MCMV-infected B6-gld/gld mice and uninfected B6-+/+ and B6-gld/gld mice. RESULTS Following MCMV infection, B6-gld/gld mice developed an acute and chronic sialadenitis characterized by multiple foci of infiltrating T cells. After local injection of adenoviral vectors, high levels of lacZ or fasL gene expression could be detected in acinar and ductal cells. Treatment of acute and chronic sialadenitis in B6-gld/gld mice with local fasL gene transfer resulted in a significant reduction in the number of inflammatory foci and tissue destruction in salivary glands compared with mice treated with AdCMVLacZ. Despite high levels of FasL expression after injection of recombinant vectors, <5% of ductal and acinar cells were TUNEL positive, demonstrating that, in this model of SS, acinar and ductal cells were not highly sensitive to FasL-mediated apoptosis. CONCLUSION Chronic sialadenitis similar to that of SS developed in B6-gld/gld mice after MCMV infection. FasL expression was reconstituted by local gene transfer, resulting in significant reduction of infiltrating mononuclear cells, which indicates that local gene transfer of fasL might be a novel treatment for chronic sialadenitis.
Collapse
Affiliation(s)
- M Fleck
- University of Regensburg, Germany
| | | | | | | | | | | |
Collapse
|
45
|
Huang JH, Tykocinski ML. CTLA-4-Fas ligand functions as a trans signal converter protein in bridging antigen-presenting cells and T cells. Int Immunol 2001; 13:529-39. [PMID: 11282992 DOI: 10.1093/intimm/13.4.529] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Co-stimulator blockade and trans inhibitory signaling, using agents such as CTLA-4-Ig and Fas ligand (FasL) respectively have been invoked as alternative strategies for suppressing pathogenic T cells. This study describes a novel hetero-bifunctional fusion protein, CTLA-4-FasL, designed to combine within a single protein both co-stimulator blocking and trans inhibitory signaling potentials. A chimeric expression cassette, in which the ectodomain coding sequences for CTLA-4 and FasL were linked in-frame, was used to produce a CTLA-4-FasL fusion protein. CTLA-4-FasL binding to both B7-1/B7-2-expressing Daudi B cells and Fas-expressing Jurkat T cells was documented by immunofluorescence and flow cytometry. The capacity of CTLA-4-FasL to induce apoptosis in Jurkat targets was markedly enhanced by the addition of Daudi and other B7-1/B7-2(+) B cell lines, which provided a membrane platform for the otherwise soluble CTLA-4-fusion protein. Moreover, in dual-chamber experiments, Daudi cells pre-coated with CTLA-4-FasL demonstrated Jurkat inhibitory activity that was cell-contact dependent. Significantly, when used to inhibit in vitro cellular proliferation of peripheral blood mononuclear cells, CTLA-4-FasL was approximately 1000-fold more potent than the extensively characterized CTLA-4-Ig fusion protein. Furthermore, the degree of inhibition induced by CTLA-4-FasL substantially surpassed that observed for CTLA-4-Ig and a soluble FasL when used in combination. CTLA-4-FasL represents the first of a novel class of fusion proteins, designated here as 'trans signal converter proteins', that combine trans signal masking and direct trans signaling functions.
Collapse
Affiliation(s)
- J H Huang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, 6 Gates Pavilion, 3400 Spruce Street, Philadelphia, PA 19104-4283, USA
| | | |
Collapse
|
46
|
Wu JM, Wu B, Miagkov A, Adams RN, Drachman DB. Specific immunotherapy of experimental myasthenia gravis in vitro: the "guided missile" strategy. Cell Immunol 2001; 208:137-47. [PMID: 11333146 DOI: 10.1006/cimm.2001.1778] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
We describe a strategy for specific immunotherapy of myasthenia gravis (MG) based on genetic engineering of antigen presenting cells (APCs) to present the autoantigen acetylcholine receptor (AChR) and express the "warhead" Fas ligand (FasL). For transduction of APCs we prepared recombinant attenuated vaccinia virus vectors carrying the following three gene constructs: (i) AChR fused to LAMP1 to present AChR and target AChR-specific T cells; (ii) FasL to eliminate the targeted T cells; and (iii) truncated FADD to protect APCs from self-destruction by FasL. The engineered APCs effectively expressed the genes of interest and killed AChR-specific T cells in culture by the Fas/FasL pathway. T cells specific for an unrelated antigen were spared. Our in vitro demonstration that engineered APCs target and kill antigen-specific T cells represents a promising novel strategy for specific immunotherapy of MG and other autoimmune diseases.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Animals
- Antigen-Presenting Cells/immunology
- Antigens, CD/genetics
- Antigens, CD/immunology
- Autoantigens/genetics
- Autoantigens/immunology
- Carrier Proteins/genetics
- Carrier Proteins/immunology
- Cell Line
- Fas Ligand Protein
- Fas-Associated Death Domain Protein
- Female
- Gene Expression
- Genetic Vectors
- Immunotherapy
- Lysosomal-Associated Membrane Protein 1
- Lysosomal Membrane Proteins
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Inbred MRL lpr
- Myasthenia Gravis, Autoimmune, Experimental/immunology
- Myasthenia Gravis, Autoimmune, Experimental/therapy
- Rats
- Rats, Inbred Lew
- Receptors, Cholinergic/genetics
- Receptors, Cholinergic/immunology
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- T-Lymphocytes/immunology
- Tumor Cells, Cultured
- Vaccinia virus
- fas Receptor/immunology
Collapse
Affiliation(s)
- J M Wu
- Neuromuscular Research Laboratory, Johns Hopkins School of Medicine, Baltimore, Maryland, 21287-7519, USA
| | | | | | | | | |
Collapse
|
47
|
Takashima A, Mummert M, Kitajima T, Matsue H. New technologies to prevent and treat contact hypersensitivity responses. Ann N Y Acad Sci 2001; 919:205-13. [PMID: 11083110 DOI: 10.1111/j.1749-6632.2000.tb06880.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Allergic contact dermatitis is a common inflammatory skin disease caused by T cells that recognize environmental and industrial allergens (i.e., haptens). Langerhans' cells (LC), which are skin-specific and "immature" members of the dendritic cell (DC) family of antigen-presenting cells, play crucial roles in the induction of contact hypersensitivity (CH) responses. Upon exposure to haptens, LC migrate from the epidermis to draining lymph nodes, mature into T cell-stimulatory DC, and activate hapten-reactive T cells. Therefore, CH responses should be preventable at the sensitization phase by interfering with one of these changes that occur in LC. Our objective is to develop new technologies for the prevention and treatment of allergic contact dermatitis. In this article, we will introduce three technologies that we have recently developed. First, using a phage display strategy, we have identified a 12-mer peptide (termed "peptide 1") that binds and blocks the function of hyaluronan (HA), which is known to serve as an adhesive substrate for LC migration. Local injection of peptide 1 in mice before topical application of DNFB blocked almost completely the emigration of LC from the epidermis to the draining lymph node, where antigen presentation takes place. Peptide 1 represents a new strategy that is designed to inhibit the initial event of CH. Second, we have established an in vitro experimental system to study the terminal maturation of LC during antigen-specific interaction with T cells. This experimental system, which employs a long-term LC line and T cell clones, should provide a unique tool for the identification of new immunosuppressive agents that block LC terminal maturation selectively. Finally, under the hypothesis that LC, which are engineered to overexpress a death ligand, would deliver apoptotic signals instead of activation signals to T cells, we created a "killer" LC clone by introducing CD95L cDNA into our long-term LC line XS106. In vivo administration of DNFB-pulsed killer LC into mice, either before or after sensitization, resulted in marked suppression of CH responses to DNFB. The killer LC technology represents an entirely new immunosuppressive therapy that is designed to eliminate only the pathogenic T cells.
Collapse
Affiliation(s)
- A Takashima
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas 75235, USA.
| | | | | | | |
Collapse
|
48
|
Watzlik A, Dufter C, Jung M, Opelz G, Terness P. Generation of FASL-expressing antigen-presenting cells for suppression of alloreactive T cells in the rat. Transplant Proc 2001; 33:599-601. [PMID: 11266975 DOI: 10.1016/s0041-1345(00)02159-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- A Watzlik
- Institute of Immunology, Department of Transplantation Immunology, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | |
Collapse
|
49
|
Abstract
Rheumatoid arthritis (RA) is a painful chronic disorder. Conventional therapies are palliative, not curative. Advances in the understanding of the pathophysiology of RA have led to the development of new therapeutic strategies, including gene therapy. Multiple studies in several different animal models provide proof supporting the use of gene therapy in arthritis. A phase I clinical trial has already been performed successfully on nine women with end-stage RA in the United States, and two other trials are in progress. Limited duration of gene expression impedes the development of a clinically useful genetic treatment for arthritis.
Collapse
Affiliation(s)
- E Gouze
- Center for Molecular Orthopaedics, Harvard Medical School, 221 Longwood Avenue, BL-152, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
50
|
Schmidt MR, Piekos B, Cabatingan MS, Woodland RT. Expression of a human coxsackie/adenovirus receptor transgene permits adenovirus infection of primary lymphocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:4112-9. [PMID: 11034423 DOI: 10.4049/jimmunol.165.7.4112] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Replication-defective adenoviruses are effective vehicles for gene transfer, both for the repair of defective genes and for studies of gene function in primary cells. Many cell types, including lymphocytes, are refractory to adenovirus infection because they lack the Coxsackie/adenovirus receptor (CAR) needed for virus attachment. To extend the advantages of adenovirus-mediated gene transfer to primary lymphoid populations and other cell types lacking endogenous CAR, we produced a mouse that expresses human (h) CAR as a transgene under control of a murine MHC class I promoter. hCAR protein is expressed on T and B lymphocytes from a variety of organs (spleen, lymph node, bone marrow, thymus, and peritoneum). These lymphocytes are susceptible to adenovirus infection, as demonstrated by reporter green fluorescent protein gene expression, with the fraction of expressing cells as high as 70%. Some lymphocyte subpopulations required stimulation subsequent to adenovirus infection for reporter expression. This activation requirement is a restriction imposed by the promoter used in the adenovirus construct. In subpopulations requiring activation, the elongation factor 1 promoter was far superior to a hCMV promoter for directing green fluorescent protein production. We also find that hCAR mRNA is produced in nonlymphoid tissues from all founder lines, including tissues that do not express endogenous murine CAR, suggesting the opportunity for effecting gene delivery to and testing gene function in a wide variety of primary cell types previously resistant to gene transfer.
Collapse
Affiliation(s)
- M R Schmidt
- Department of Molecular Genetics and Microbiology, University of Massachusetts Medical School, Worcester 01655, USA.
| | | | | | | |
Collapse
|