51
|
Hu P, Wang G, Wu S, Zhu H, Ping L, Wang C, Shen G. Transplantation of NIT-1 cells with ectopic FADDdel-GFP expression for treatment of streptozotocin-induced diabetes. Autoimmunity 2009; 42:424-31. [PMID: 19811259 DOI: 10.1080/08916930902887078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Islet transplantation is considered a therapeutic option for type 1 diabetes (T1D). However, allorejection is one major barrier for the successful islet transplantation. In the present study, we have tested the feasibility of a deletion construct for Fas-associated death domain protein (FADD; without the death effecter domain) fused with green fluorescent protein (FADDdel-GFP) for blocking the Fas-FasL signaling pathway in prevention of transplanted beta cell destruction by allo-rejection in T1D. In vitro studies have shown that NIT-1 cells with ectopic FADDdel expression were resistant to cytokine-induced apoptosis and CTL-mediated lysis. Diabetic Balb/c mice reached normoglycemia promptly and gained weight after transplantation of NIT-1 cells with ectopic FADDdel-GFP expression. These recipients showed a significant longer survival time than that of recipients transplanted with NIT cells with ectopic GFP expression only. Our results together suggest that FADDdel could be a useful target for the improvement of islet transplantation for T1D.
Collapse
Affiliation(s)
- Ping Hu
- Department of Microbiology and Immunology, Medical College of Jinan University, Guangzhou, 510632, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
52
|
Dirice E, Sanlioglu AD, Kahraman S, Ozturk S, Balci MK, Omer A, Griffith TS, Sanlioglu S. Adenovirus-Mediated TRAIL Gene (Ad5hTRAIL) Delivery into Pancreatic Islets Prolongs Normoglycemia in Streptozotocin-Induced Diabetic Rats. Hum Gene Ther 2009; 20:1177-89. [DOI: 10.1089/hum.2009.039] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Ercument Dirice
- Human Gene Therapy Division, Department of Medical Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
- Department of Medical Biology and Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
| | - Ahter Dilsad Sanlioglu
- Human Gene Therapy Division, Department of Medical Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
- Department of Medical Biology and Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
| | - Sevim Kahraman
- Human Gene Therapy Division, Department of Medical Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
- Department of Medical Biology and Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
| | - Saffet Ozturk
- Department of Histology and Embryology, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
| | - Mustafa Kemal Balci
- Division of Endocrinology and Metabolic Diseases, Department of Medicine, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
| | - Abdulkadir Omer
- Section on Islet Transplantation and Cell Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, U.S.A
| | | | - Salih Sanlioglu
- Human Gene Therapy Division, Department of Medical Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
- Department of Medical Genetics, Faculty of Medicine, Akdeniz University, Antalya 07070, Turkey
| |
Collapse
|
53
|
Cotransplantation with xenogenetic neonatal porcine sertoli cells significantly prolongs islet allograft survival in nonimmunosuppressive rats. Transplantation 2009; 88:339-45. [PMID: 19667935 DOI: 10.1097/tp.0b013e3181ae5dcf] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND In addition to possessing immune privileged properties, Sertoli cells are known to actively suppress responses to cotransplanted cells. An important question is whether this "bystander suppression" is limited to cells of the same origin as the Sertoli cells or whether suppression extends to unrelated cells. METHODS Neonatal porcine Sertoli cells (NPSCs) were transplanted with allogeneic islets (Sprague-Dawley rat) into immune competent Wistar rats subsequent to induction of diabetes by alloxan administration. RESULTS Although allogeneic islets alone had a mean survival time of 5.67+/-0.94 days, islets cotransplanted with 1.5 x 10 xenogeneic NPSCs displayed a survival of 8.33+/-0.58 days. Increasing the concentration of NPSCs to 1.0 x10 yielded a further increase in survival to 16.33+/-1.53 days. Augmented islet survival was associated with reduced lymphocytic infiltrate and elevated numbers of Sox9 positive cells. Mechanistically, it seemed that Fas ligand was not involved in prolongation of survival because in contrast to adult Sertoli cells, NPSCs lacked expression of this gene. CONCLUSIONS These data suggest that xenogeneic Sertoli cells exert a global immune suppressive effect that extends across species barriers in a stringent model of alloimmune rejection. The combination of NPSCs with other immune modulatory regimes may yield novel approaches toward prevention of allo-islet transplant rejection.
Collapse
|
54
|
A placental protective role for trophoblast-derived TNF-related apoptosis-inducing ligand (TRAIL). Placenta 2009; 30:855-60. [PMID: 19674787 DOI: 10.1016/j.placenta.2009.07.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2009] [Revised: 07/14/2009] [Accepted: 07/16/2009] [Indexed: 11/22/2022]
Abstract
Recent studies show that apoptosis, programmed cell death, plays an important role in the normal development of the human placenta and that an altered balance between proliferation and apoptosis of villous trophoblasts is associated with abnormal pregnancies. The TNF-related apoptosis-inducing ligand (TRAIL) is a molecule belonging to TNF superfamily. The role of TRAIL and its Death Receptor 5 (DR5) in regulating villous trophoblast cell turnover in normal and pathologic pregnancies remains to be explored. In order to elucidate the role of TRAIL in the regulation of placental growth, primary cytotrophoblast cells were isolated from normal term placentas (n=13) and cultured for 18 and 66h to generate mononucleate and multinucleate trophoblasts, respectively. The protein expression and localisation of TRAIL and DR5 were determined by Western blotting and immunofluorescence. Secreted sTRAIL was also measured by ELISA. Trophoblast apoptosis was measured by TUNEL in the presence of recombinant TRAIL (rTRAIL), and DR5 relocalisation was assessed by immunostaining after 18h exposure to TNFalpha. We demonstrated that TRAIL protein expression and the secretion of soluble TRAIL (sTRAIL) were down-regulated in syncytialised villous trophoblasts and that sTRAIL was independent of biochemical differentiation, as TRAIL-neutralizing antibody (2E5) failed to influence hCG production. TRAIL immunoreactivity was detected in mono- and multinucleated trophoblast cells and localised to the cytoplasm and cellular membranes -- more intense staining was associated with apoptotic nuclei. rTRAIL failed to induce apoptosis in trophoblasts cells owing to the nuclear localisation of DR5. However, TNFalpha treatment caused the redistribution of intracellular DR5 to the cell surface, potentiating apoptotic susceptibly to exogenously administered rTRAIL. These findings highlight a mechanism by which TRAIL and DR5 serve to protective trophoblasts in normal development, but may be activated in conditions of excessive TNFalpha.
Collapse
|
55
|
Cheng X, Dai H, Wan N, Moore Y, Vankayalapati R, Dai Z. Interaction of programmed death-1 and programmed death-1 ligand-1 contributes to testicular immune privilege. Transplantation 2009; 87:1778-86. [PMID: 19543053 DOI: 10.1097/tp.0b013e3181a75633] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Immune responses are tempered in immunologically privileged sites including the testis. Previous studies have shown that islet transplantation in the testis significantly prolongs islet allograft survival. However, mechanisms underlying testicular immune privilege and intratesticular allograft survival remain unclear. METHODS Allogeneic murine islets were transplanted in the testis. Programmed death-1 ligand-1 (PD-L1) expression was detected by immunohistochemstry and real-time polymerase chain reaction. Infiltrating T-cell proliferation was measured by bromodeoxyuridine uptakes, whereas their apoptosis was quantified by terminal deoxynucleotide transferase-mediated dUTP nick-end labeling methods. Transgenic T cells were used to track allospecific memory T-cell generation. RESULTS We found that programmed death-1 (PD-1):PD-L1 negative costimulation is essential for prolonged survival of intratesticular islet allografts, as blocking PD-L1 or PD-1, but not PD-L2 and cytotoxic T-lymphocyte antigen 4, abrogated long-term survival of intratesticular islet allografts. As controls, blocking PD-1 or PD-L1 did not significantly accelerate the acute rejection of islet allografts transplanted under the renal capsule, a conventional islet-grafting site. We also found for the first time that PD-L1 is constitutively expressed mainly by spermatocytes and spermatids in seminiferous tubules of the testis. Moreover, infiltrating T cells underwent less vigorous proliferation but faster apoptosis in the testis than in the kidney. Blocking PD-1:PD-L1 costimulation largely abolished the suppression of T-cell proliferation and acceleration of T-cell apoptosis. Importantly, testicular immune privilege significantly suppressed the generation and proliferation of donor-specific memory CD8 T cells. CONCLUSIONS The constitutive expression of PD-L1 in the testis is an important mechanism underlying testicular immune privilege and long-term survival of intratesticular islet allografts.
Collapse
Affiliation(s)
- Xuyang Cheng
- Center for Biomedical Research, University of Texas Health Science Center, Tyler, TX 75708, USA
| | | | | | | | | | | |
Collapse
|
56
|
Laurence JM, Allen RDM, McCaughan GW, Logan GJ, Alexander IE, Bishop GA, Sharland AF. Gene therapy in transplantation. Transplant Rev (Orlando) 2009; 23:159-70. [PMID: 19428235 DOI: 10.1016/j.trre.2009.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Gene therapy is an exciting and novel technology that offers the prospect of improving transplant outcomes beyond those achievable with current clinical protocols. This review explores both the candidate genes and ways in which they have been deployed to overcome both immune and non-immune barriers to transplantation success in experimental models. Finally, the major obstacles to implementing gene therapy in the clinic are considered.
Collapse
Affiliation(s)
- Jerome M Laurence
- Collaborative Transplantation Research Group, Bosch Insitute, Royal Prince Alfred Hospital and University of Sydney, NSW 2006, Australia
| | | | | | | | | | | | | |
Collapse
|
57
|
Diabetes mellitus and apoptosis: inflammatory cells. Apoptosis 2009; 14:1435-50. [PMID: 19360474 DOI: 10.1007/s10495-009-0340-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Accepted: 03/17/2009] [Indexed: 12/24/2022]
Abstract
Since the early observation that similarities between thyroiditis and insulitis existed, the important role played by inflammation in the development of diabetes has been appreciated. More recently, experiments have shown that inflammation also plays a prominent role in the development of target organ damage arising as complications, with both elements of the innate and the adaptive immune system being involved, and that cytokines contributing to local tissue damage may arise from both infiltrating and resident cells. This review will discuss the experimental evidence that shows that inflammatory cell-mediated apoptosis contributes to target organ damage, from beta cell destruction to both micro- and macro-vascular disease complications, and also how alterations in leukocyte turnover affects immune function.
Collapse
|
58
|
Molinero LL, Wang Y, Zhou P, Yagita H, Alegre ML. Fas mediates cardiac allograft acceptance in mice with impaired T-cell-intrinsic NF-kappaB signaling. Transpl Int 2009; 22:845-52. [PMID: 19351347 DOI: 10.1111/j.1432-2277.2009.00875.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The transcription factor NF-kappaB is critical for T-cell activation and survival. We have shown that mice expressing a T-cell-restricted NF-kappaB superrepressor (IkappaBalphaDeltaN-Tg) permanently accept heart but not skin allografts. Overexpression of the prosurvival factor Bcl-x(L) in T cells restored heart rejection, suggesting that graft acceptance in IkappaBalphaDeltaN-Tg mice was attributable to deletion of alloreactive T cells.In vitro, the increased death of IkappaBalphaDeltaN-Tg T cells upon TCR stimulation when compared with wildtype T cells was mostly because of Fas/FasL interaction. Similarly, Fas played a key role in cardiac allograft acceptance by IkappaBalphaDeltaN-Tg mice as both genetic and antibody-mediated inhibition of Fas-signaling restored cardiac allograft rejection. Rejection correlated with graft infiltration by T cells and splenic production of IFN-gamma upon allostimulation. These results indicate that T-cell inhibition of NF-kappaB results in cardiac allograft acceptance because of increased susceptibility to Fas-mediated cell death.
Collapse
Affiliation(s)
- Luciana Lorena Molinero
- Department of Medicine, Section of Rheumatology, The University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA.
| | | | | | | | | |
Collapse
|
59
|
Abstract
FAS belongs to the subgroup of the tumor necrosis factor receptor (TNF-R) family that contains an intracellular "death domain" and triggers apoptosis. Its physiological ligand FASL is a member of the TNF cytokine family. Studies with mutant mice and cells from human patients have shown that FAS plays critical roles in the immune system, including the killing of pathogen-infected cells and the death of obsolete and potentially dangerous lymphocytes. Fas thereby functions as a guardian against autoimmunity and tumor development. FAS triggers apoptosis through FADD-mediated recruitment and activation of caspase-8. In certain cells such as hepatocytes, albeit not lymphocytes, FAS-induced apoptosis requires amplification through proteolytic activation of the proapoptotic BCL-2 family member BID. Curiously, several components of the FAS signaling machinery have been implicated in nonapoptotic processes, including cellular activation, differentiation, and proliferation. This review describes current understanding of Fas-induced apoptosis signaling and proposes experimental strategies for future advances.
Collapse
Affiliation(s)
- Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.
| | | | | |
Collapse
|
60
|
Ramaswamy M, Cleland SY, Cruz AC, Siegel RM. Many checkpoints on the road to cell death: regulation of Fas-FasL interactions and Fas signaling in peripheral immune responses. Results Probl Cell Differ 2009; 49:17-47. [PMID: 19132321 DOI: 10.1007/400_2008_24] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Interactions between the TNF-family receptor Fas (CD95) and Fas Ligand (FasL, CD178) can efficiently induce apoptosis and are critical for the maintenance of immunological self-tolerance. FasL is kept under strict control by transcriptional and posttranslational regulation. Surface FasL can be cleaved by metalloproteases, resulting in shed extracellular domains, and FasL can also traffic to secretory lysosomes. Each form of FasL has distinct biological functions. Fas is more ubiquitously expressed, but its apoptosis-inducing function is regulated by a number of mechanisms including submembrane localization, efficiency of receptor signaling complex assembly and activation, and bcl-2 family members in some circumstances. When apoptosis is not induced, Fas-FasL interactions can also trigger a number of activating and proinflammatory signals. Harnessing the apoptosis-inducing potential of Fas for therapy of cancer and autoimmune disease has been actively pursued, and despite a number of unexpected side-effects that result from manipulating Fas-FasL interactions, this remains a worthy goal.
Collapse
Affiliation(s)
- Madhu Ramaswamy
- Immunoregulation Unit, Autoimmunity Branch, NIAMS, National Institutes of Health, Bethesda MD 20892, USA
| | | | | | | |
Collapse
|
61
|
Kawamoto K, Tanemura M, Ito T, Deguchi T, Machida T, Nishida T, Doki Y, Mori M, Sawa Y. Prolonged survival of pig islets xenograft by adenovirus-mediated expression of either the membrane-bound human FasL or the human decoy Fas antigen gene. Xenotransplantation 2008; 15:333-43. [DOI: 10.1111/j.1399-3089.2008.00490.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
62
|
Yolcu ES, Gu X, Lacelle C, Zhao H, Bandura-Morgan L, Askenasy N, Shirwan H. Induction of tolerance to cardiac allografts using donor splenocytes engineered to display on their surface an exogenous fas ligand protein. THE JOURNAL OF IMMUNOLOGY 2008; 181:931-9. [PMID: 18606644 DOI: 10.4049/jimmunol.181.2.931] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The critical role played by Fas ligand (FasL) in immune homeostasis renders this molecule an attractive target for immunomodulation to achieve tolerance to auto- and transplantation Ags. Immunomodulation with genetically modified cells expressing FasL was shown to induce tolerance to alloantigens. However, genetic modification of primary cells in a rapid, efficient, and clinically applicable manner proved challenging. Therefore, we tested the efficacy of donor splenocytes rapidly and efficiently engineered to display on their surface a chimeric form of FasL protein (SA-FasL) for tolerance induction to cardiac allografts. The i.p. injection of ACI rats with Wistar-Furth rat splenocytes displaying SA-FasL on their surface resulted in tolerance to donor, but not F344 third-party cardiac allografts. Tolerance was associated with apoptosis of donor reactive T effector cells and induction/expansion of CD4(+)CD25(+)FoxP3(+) T regulatory (Treg) cells. Treg cells played a critical role in the observed tolerance as adoptive transfer of sorted Treg cells from long-term graft recipients into naive unmanipulated ACI rats resulted in indefinite survival of secondary Wistar-Furth grafts. Immunomodulation with allogeneic cells rapidly and efficiently engineered to display on their surface SA-FasL protein provides an effective and clinically applicable means of cell-based therapy with potential application to regenerative medicine, transplantation, and autoimmunity.
Collapse
Affiliation(s)
- Esma S Yolcu
- Department of Microbiology and Immunology, Institute for Cellular Therapeutics, University of Louisville, Louisville, KY 40202, USA.
| | | | | | | | | | | | | |
Collapse
|
63
|
Fang Y, Braley-Mullen H. Cultured murine thyroid epithelial cells expressing transgenic Fas-associated death domain-like interleukin-1beta converting enzyme inhibitory protein are protected from fas-mediated apoptosis. Endocrinology 2008; 149:3321-9. [PMID: 18356280 PMCID: PMC2453085 DOI: 10.1210/en.2008-0080] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The antiapoptotic molecule Fas-associated death domain-like IL-1beta-converting enzyme inhibitory protein (FLIP) inhibits Fas-mediated apoptosis by blocking activation of caspase-8. We previously showed that expression of transgenic FLIP on thyroid epithelial cells (TECs) of DBA/1 and CBA/J mice promoted earlier resolution of granulomatous experimental autoimmune thyroiditis in vivo. This study was undertaken to directly determine whether transgenic FLIP expressed on cultured TECs can protect TECs from Fas-mediated apoptosis in vitro. The results indicate that cultured TECs from DBA/1 and CBA/J mice can be sensitized in vitro by interferon-gamma and TNF-alpha to undergo Fas-mediated apoptosis. Transgenic overexpression of FLIP protected cultured TECs of FLIP transgene (Tg)+ DBA/1 and CBA/J mice from Fas-mediated apoptosis, and FLIP small interfering RNA transfection of cultured TECs of FLIP Tg+ DBA/1 and CBA/J mice abolished the protective effect. These in vitro results are consistent with our previous in vivo studies using DBA/1 and CBA/J FLIP Tg+ mice and provide direct support for the hypothesis that transgenic expression of FLIP promotes resolution of granulomatous experimental autoimmune thyroiditis by protecting TECs from apoptosis.
Collapse
Affiliation(s)
- Yujiang Fang
- Division of Immunology and Rheumatology, Department of Medicine, University of Missouri, NE307 Medical Sciences, Columbia, MO 65212, USA.
| | | |
Collapse
|
64
|
Buonocore S, Haddou NO, Moore F, Florquin S, Paulart F, Heirman C, Thielemans K, Goldman M, Flamand V. Neutrophil-dependent tumor rejection and priming of tumoricidal CD8+ T cell response induced by dendritic cells overexpressing CD95L. J Leukoc Biol 2008; 84:713-20. [PMID: 18567840 DOI: 10.1189/jlb.0108075] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Overexpression of CD95 (Fas/Apo-1) ligand (CD95L) has been shown to induce T cell tolerance but also, neutrophilic inflammation and rejection of allogeneic tissue. We explored the capacity of dendritic cells (DCs) genetically engineered to overexpress CD95L to induce an antitumor response. We first found that DCs overexpressing CD95L, in addition to MHC class I-restricted OVA peptides (CD95L-OVA-DCs), induced increased antigen-specific CD8(+) T cell responses as compared with DCs overexpressing OVA peptides alone. The enhanced T cell responses were associated with improved regression of a tumor expressing OVA, allowing survival of all animals. When DCs overexpressing CD95L (CD95L-DCs) were injected with the tumor expressing OVA, in vivo tumor proliferation was strikingly inhibited. A strong cellular apoptosis and a massive neutrophilic infiltrate developed in this setting. Neutrophil depletion prevented tumor regression as well as enhanced IFN-gamma production induced by CD95L-OVA-DCs. Furthermore, the CD8(+) T cell response induced by the coadministration of tumor cells and CD95L-DCs led to rejection of a tumor implanted at a distance from the DC injection site. In summary, DCs expressing CD95L promote tumor rejection involving neutrophil-mediated innate immunity and CD8(+) T cell-dependent adaptative immune responses.
Collapse
Affiliation(s)
- Sofia Buonocore
- Institute for Medical Immunology, Université Libre de Bruxelles, Rue Adrienne Bolland 8, 6041 Gosselies, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Wilson JT, Chaikof EL. Challenges and emerging technologies in the immunoisolation of cells and tissues. Adv Drug Deliv Rev 2008; 60:124-45. [PMID: 18022728 DOI: 10.1016/j.addr.2007.08.034] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2007] [Accepted: 08/13/2007] [Indexed: 12/22/2022]
Abstract
Protection of transplanted cells from the host immune system using immunoisolation technology will be important in realizing the full potential of cell-based therapeutics. Microencapsulation of cells and cell aggregates has been the most widely explored immunoisolation strategy, but widespread clinical application of this technology has been limited, in part, by inadequate transport of nutrients, deleterious innate inflammatory responses, and immune recognition of encapsulated cells via indirect antigen presentation pathways. To reduce mass transport limitations and decrease void volume, recent efforts have focused on developing conformal coatings of micron and submicron scale on individual cells or cell aggregates. Additionally, anti-inflammatory and immunomodulatory capabilities are being integrated into immunoisolation devices to generate bioactive barriers that locally modulate host responses to encapsulated cells. Continued exploration of emerging paradigms governed by the inherent challenges associated with immunoisolation will be critical to actualizing the clinical potential of cell-based therapeutics.
Collapse
|
66
|
Fang Y, DeMarco VG, Sharp GC, Braley-Mullen H. Expression of transgenic FLIP on thyroid epithelial cells inhibits induction and promotes resolution of granulomatous experimental autoimmune thyroiditis in CBA/J mice. Endocrinology 2007; 148:5734-45. [PMID: 17823262 DOI: 10.1210/en.2007-0939] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Granulomatous experimental autoimmune thyroiditis (G-EAT) is induced by transfer of thyroglobulin-primed in vitro activated splenocytes. Thyroid lesions reach maximal severity 20 d later, and inflammation resolves or progresses to fibrosis by d 60, depending on the extent of thyroid damage at d 20. Depletion of CD8+ T cells inhibits G-EAT resolution. We showed that expression of Fas-associated death domain-like IL-1beta-converting enzyme inhibitory protein (FLIP) transgene (Tg) on thyroid epithelial cells (TECs) of DBA/1 mice had no effect on G-EAT induction but promoted earlier resolution of G-EAT. However, when CBA/J wild-type donor cells were transferred to transgenic CBA/J mice expressing FLIP on TECs, they developed less severe G-EAT than FLIP Tg- littermates. Both strains expressed similar levels of the FLIP Tg, but endogenous FLIP was up-regulated to a greater extent on infiltrating T cells during G-EAT development in DBA/1 compared with CBA/J mice. After transient depletion of CD8+ T cells, FLIP Tg+ and Tg- CBA/J recipients both developed severe G-EAT at d 20. Thyroid lesions in CD8-depleted Tg+ recipients were resolving by d 60, whereas lesions in Tg- littermates did not resolve, and most were fibrotic. FLIP Tg+ recipients had increased apoptosis of CD3+ T cells compared with Tg- recipients. The results indicate that transgenic FLIP expressed on TECs in CBA/J mice promotes G-EAT resolution, but induction of G-EAT is inhibited unless CD8+ T cells are transiently depleted.
Collapse
MESH Headings
- Animals
- Apoptosis/immunology
- Blotting, Western
- CASP8 and FADD-Like Apoptosis Regulating Protein/genetics
- CASP8 and FADD-Like Apoptosis Regulating Protein/immunology
- CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism
- CD3 Complex/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cytokines/immunology
- Disease Models, Animal
- Epithelial Cells/immunology
- Epithelial Cells/metabolism
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/immunology
- Forkhead Transcription Factors/metabolism
- Mice
- Mice, Inbred CBA
- Mice, Transgenic
- Microscopy, Confocal
- Reverse Transcriptase Polymerase Chain Reaction
- Spleen/cytology
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- Thyroid Gland/cytology
- Thyroiditis, Autoimmune/genetics
- Thyroiditis, Autoimmune/immunology
- Thyroiditis, Autoimmune/metabolism
- Thyroiditis, Subacute/genetics
- Thyroiditis, Subacute/immunology
- Thyroiditis, Subacute/metabolism
Collapse
Affiliation(s)
- Yujiang Fang
- Division of Immunology and Rheumatology, Department of Medicine, University of Missouri, NE307 Medical Sciences, Columbia, MO 65212, USA
| | | | | | | |
Collapse
|
67
|
Kuttler B, Wanka H, Klöting N, Gerstmayer B, Volk HD, Sawitzki B, Ritter T. Ex vivo gene transfer of viral interleukin-10 to BB rat islets: no protection after transplantation to diabetic BB rats. J Cell Mol Med 2007; 11:868-80. [PMID: 17760846 PMCID: PMC3823263 DOI: 10.1111/j.1582-4934.2007.00059.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Allogeneic and autoimmune islet destruction limits the success of islet transplantation in autoimmune diabetic patients. This study was designed to investigate whether ex vivo gene transfer of viral interleukin-10 (vIL-10) protects BioBreeding (BB) rat islets from autoimmune destruction after transplantation into diabetic BB recipients. Islets were transduced with adenoviral constructs (Ad) expressing the enhanced green fluorescent protein (eGFP), α-1 antitrypsin (AAT) or vIL-10. Transduction efficiency was demonstrated by eGFP-positive cells and vIL-10 production. Islet function was determined in vitro by measuring insulin content and insulin secretion and in vivo by grafting AdvIL-10-transduced islets into syngeneic streptozotocin (SZ)-diabetic, congenic Lewis (LEW.1 W) rats. Finally, gene-modified BB rat islets were grafted into autoimmune diabetic BB rats. Ad-transduction efficiency of islets increased with virus titre and did not interfere with insulin content and insulin secretion. Ad-transduction did not induce Fas on islet cells. AdvIL-10-transduced LEW.1 W rat islets survived permanently in SZ-diabetic LEW.1 W rats. In diabetic BB rats AdvIL-10-transduced BB rat islets were rapidly destroyed. Prolongation of islet culture prior to transplantation improved the survival of gene-modified islets in BB rats. Several genes including those coding for chemokines and other peptides associated with inflammation were down-regulated in islets after prolonged culture, possibly contributing to improved islet graft function in vivo. Islets transduced ex vivo with vIL-10 are principally able to cure SZ-diabetic rats. Autoimmune islet destruction in diabetic BB rats is not prevented by ex vivo vIL-10 gene transfer to grafted islets. Graft survival in autoimmune diabetic rats may be enhanced by improvements in culture conditions prior to transplantation.
Collapse
Affiliation(s)
- Beate Kuttler
- Institute of Physiology, Medical School, Ernst-Moritz-Arndt-University of Greifswald, Greifswalderstrasse 11c, D-17495 Karlsburg, Germany.
| | | | | | | | | | | | | |
Collapse
|
68
|
Fujimori K, Oh-i K, Takeuchi M, Yamakawa N, Hattori T, Kezuka T, Keino H, Suzuki J, Goto H, Sakai JI, Usui M. Circulating neutrophils in Behçet disease is resistant for apoptotic cell death in the remission phase of uveitis. Graefes Arch Clin Exp Ophthalmol 2007; 246:285-90. [PMID: 17690898 DOI: 10.1007/s00417-007-0659-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2007] [Revised: 07/05/2007] [Accepted: 07/08/2007] [Indexed: 10/23/2022] Open
Abstract
BACKGROUND Behçet disease (BD) is manifested by recurrent acute iridocyclitis with hypopyon in the active phase, which regresses spontaneously. Hypopyon consists of inflammatory cells infiltrating the eye, with polymorphonuclear cells (PMNs) as the main component. The present study was conducted to investigate the apoptosis property of PMNs in BD patients with uveitis. METHODS PMNs were purified from peripheral blood cells of BD patients with uveitis in the active or remission phase and were cultured for 12 hours. In some cultures, lipopolysaccharide (LPS), antagonistic anti-TNFalpha antibody, agonistic anti-Fas antibody, or Fas:Fc fusion protein was added. At the end of cultures, apoptotic cells were evaluated by Annexin V expression using flow cytometry. RESULTS Spontaneous apoptosis of PMNs showed lower levels in the remission phase of BD-related uveitis compared with the active phase or healthy controls. The lower level of PMN apoptosis in the remission phase of uveitis in BD remained even by stimulation with LPS, anti-TNFalpha antibody, or Fas:Fc fusion protein, which was abolished in the presence of agonistic anti-Fas antibody. CONCLUSIONS In BD patients, the apoptosis of PMNs was reduced in the remission phase of uveitis and restored in the active phase, which arose from the apoptotic cell death in part via Fas-Fas ligand interaction.
Collapse
Affiliation(s)
- Keita Fujimori
- Department of Ophthalmology, Tokyo Medical University, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Kaminitz A, Stein J, Yaniv I, Askenasy N. The vicious cycle of apoptotic beta-cell death in type 1 diabetes. Immunol Cell Biol 2007; 85:582-9. [PMID: 17637698 DOI: 10.1038/sj.icb.7100093] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Autoimmune insulitis, the cause of type 1 diabetes, evolves through several discrete stages that culminate in beta-cell death. In the first stage, antigenic epitopes of B-cell-specific peptides are processed by antigen presenting cells in local lymph nodes, and auto-reactive lymphocyte clones are propagated. Subsequently, cell-mediated and direct cytokine-mediated reactions are generated against the beta-cells, and the beta-cells are sensitized to apoptosis. Ironically, the beta-cells themselves contribute some of the cytokines and chemokines that provoke the immune reaction within the islets. Once this vicious cycle of autoimmunity is fully developed, the fate of the beta-cells in the islets is sealed, and clinical diabetes inevitably ensues. Differences in various aspects of these concurrent events appear to underlie the significant discrepancies in experimental data observed in experimental models that simulate autoimmune insulitis.
Collapse
Affiliation(s)
- Ayelet Kaminitz
- Frankel Laboratory, Center for Stem Cell Research, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | | | | | | |
Collapse
|
70
|
Altemeier WA, Zhu X, Berrington WR, Harlan JM, Liles WC. Fas (CD95) induces macrophage proinflammatory chemokine production via a MyD88-dependent, caspase-independent pathway. J Leukoc Biol 2007; 82:721-8. [PMID: 17576821 PMCID: PMC4492281 DOI: 10.1189/jlb.1006652] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Activation of the prototypical death receptor, Fas (CD95), can induce both caspase-dependent cell death and production of proinflammatory chemokines, leading to neutrophil recruitment and end-organ injury. The precise mechanism(s) by which Fas up-regulates chemokine production and release, is currently unclear. We hypothesized that Fas-induced chemokine release by macrophages is dependent on the MyD88 adaptor molecule and independent of caspase activity. To test this hypothesis, we measured chemokine response to Fas activation both in RAW 264.7 cells with RNAi-attenuated MyD88 expression and in MyD88-deficient primary macrophages. We found that Fas-induced chemokine release was abrogated in the absence of MyD88. In vivo, MyD88(-/-) mice had impaired CXCL1/KC release and polymorphonuclear cell recruitment in response to intratracheal treatment with the Fas-activating monoclonal antibody, Jo-2. Furthermore, Fas-induced chemokine release was not dependent on either IL-1 receptor signaling or on caspase activity. We conclude that MyD88 plays an integral role in Fas-induced macrophage-mediated inflammation.
Collapse
Affiliation(s)
- William A Altemeier
- Department of Medicine, University of Washington School of Medicine, 1959 NE Pacific St., Seattle, WA 98105-6522, USA.
| | | | | | | | | |
Collapse
|
71
|
Fang Y, Wei Y, Demarco V, Chen K, Sharp GC, Braley-Mullen H. Murine FLIP transgene expressed on thyroid epithelial cells promotes resolution of granulomatous experimental autoimmune thyroiditis in DBA/1 mice. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 170:875-87. [PMID: 17322373 PMCID: PMC1864882 DOI: 10.2353/ajpath.2007.060816] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Granulomatous experimental autoimmune thyroiditis (G-EAT) is induced by mouse thyroglobulin-sensitized splenocytes activated in vitro with mouse thyroglobulin and interleukin-12. In wild-type (WT) DBA/1 recipients of WT donor splenocytes, thyroid lesions reach maximal severity at day 20, with ongoing inflammation and extensive fibrosis at day 60. Our previous studies indicated the site of expression of FLIP and Fas ligand [thyroid epithelial cells (TECs) versus inflammatory cells] differed in mice when lesions would resolve or progress to fibrosis. To test the hypothesis that expression of FLIP by TECs would promote earlier G-EAT resolution in DBA/1 mice, transgenic (Tg) DBA/1 mice expressing FLIP on TECs were generated. In FLIP Tg(+) and Tg(-) littermate recipients of WT donor splenocytes, G-EAT severity was comparable at day 20, but fibrosis was decreased, and many lesions resolved by day 60 in Tg(+) but not Tg(-) recipients. FLIP and Fas ligand were primarily expressed by TECs in Tg(+) recipients and by inflammatory cells in Tg(-) recipients at day 60. Apoptosis of inflammatory cells was greater, and expression of proinflammatory cytokines was decreased in thyroids of Tg(+) compared with Tg(-) recipients. These results are consistent with the hypothesis that transgenic expression of FLIP on thyroid epithelial cells promotes earlier resolution of granulomatous experimental autoimmune thyroiditis.
Collapse
Affiliation(s)
- Yujiang Fang
- Department of Internal Medicine, University of Missouri School of Medicine, and Veterans Affairs Research Service, Columbia, Missouri 65212, USA
| | | | | | | | | | | |
Collapse
|
72
|
Matsumoto N, Imamura R, Suda T. Caspase-8- and JNK-dependent AP-1 activation is required for Fas ligand-induced IL-8 production. FEBS J 2007; 274:2376-84. [PMID: 17403042 DOI: 10.1111/j.1742-4658.2007.05772.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Despite a dogma that apoptosis does not induce inflammation, Fas ligand (FasL), a well-known death factor, possesses pro-inflammatory activity. For example, FasL induces nuclear factor kappaB (NF-kappaB) activity and interleukin 8 (IL-8) production by engagement of Fas in human cells. Here, we found that a dominant negative mutant of c-Jun, a component of the activator protein-1 (AP-1) transcription factor, inhibits FasL-induced AP-1 activity and IL-8 production in HEK293 cells. Selective inhibition of AP-1 did not affect NF-kappaB activation and vice versa, indicating that their activations were not sequential events. The FasL-induced AP-1 activation could be inhibited by deleting or introducing the lymphoproliferation (lpr)-type point mutation into the Fas death domain (DD), knocking down the Fas-associated DD protein (FADD), abrogating caspase-8 expression with small interfering RNAs, or using inhibitors for pan-caspase and caspase-8 but not caspase-1 or caspase-3. Furthermore, wildtype, but not a catalytically inactive mutant, of caspase-8 reconstituted the FasL-induced AP-1 activation in caspase-8-deficient cells. Fas ligand induced the phosphorylation of two of the three major mitogen-activated protein kinases (MAPKs): extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) but not p38 MAPK. Unexpectedly, an inhibitor for JNK but not for MAPK/ERK kinase inhibited the FasL-induced AP-1 activation and IL-8 production. These results demonstrate that FasL-induced AP-1 activation is required for optimal IL-8 production, and this process is mediated by FADD, caspase-8, and JNK.
Collapse
Affiliation(s)
- Norihiko Matsumoto
- Division of Immunology and Molecular Biology, Cancer Research Institute, Kanazawa University, Japan
| | | | | |
Collapse
|
73
|
Lee HM, Oh BC, Lim DP, Lee DS, Cho J, Lee G, Lee JR. Role of complement regulatory proteins in the survival of murine allo-transplanted Sertoli cells. J Korean Med Sci 2007; 22:277-82. [PMID: 17449937 PMCID: PMC2693595 DOI: 10.3346/jkms.2007.22.2.277] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Sertoli cells (SC) are known to contain immunoprotective properties, which allow them to survive as allografts without the use of immunosuppressive drugs. Experiments were designed to determine which factors are related to prolonged survival of allogeneic SC. Balb/c derived Sertoli (TM4) and colon cancer (CT-26) cell lines were implanted beneath the kidney capsule of non-immunosuppressed C57BL/6 mice and compared their survival as allografts. Compared to TM4 graft, which survived more than 7 days after transplantation, CT-26 showed massive infiltration of polymorphonuclear cells, necrosis and enlargement of draining lymph nodes. Cultured cell lines showed no differences in their expression patterns of FasL, TGF beta1, clusterin and two complement regulatory proteins (CRP, i.e., membrane cofactor protein, MCP; decay accelerating factor, DAF), but protectin (CD59), another member of CRP was expressed only on TM4. These results suggest that CD59 and unknown factors may contribute to the prolonged survival of SC in non-immunoprivileged sites.
Collapse
Affiliation(s)
- Hak-Mo Lee
- Department of Thoracic and Cardiovascular Surgery, Cancer Research Institute, College of Medicine, Seoul, Korea
- Xenotransplantation Research Center, Seoul National University, Seoul, Korea
| | - Byoung Chol Oh
- Department of Thoracic and Cardiovascular Surgery, Cancer Research Institute, College of Medicine, Seoul, Korea
- Xenotransplantation Research Center, Seoul National University, Seoul, Korea
| | - Dong-Pyo Lim
- Department of Thoracic and Cardiovascular Surgery, Cancer Research Institute, College of Medicine, Seoul, Korea
- Xenotransplantation Research Center, Seoul National University, Seoul, Korea
| | - Dong-Sup Lee
- Department of Laboratory of Immunology, Cancer Research Institute, College of Medicine, Seoul, Korea
- Xenotransplantation Research Center, Seoul National University, Seoul, Korea
| | - Jaejin Cho
- Laboratory of Stem Cell Differentiation, College of Dentistry, Seoul, Korea
- Xenotransplantation Research Center, Seoul National University, Seoul, Korea
| | - Gene Lee
- Laboratory of Stem Cell Differentiation, College of Dentistry, Seoul, Korea
- Xenotransplantation Research Center, Seoul National University, Seoul, Korea
| | - Jeong Ryul Lee
- Department of Thoracic and Cardiovascular Surgery, Cancer Research Institute, College of Medicine, Seoul, Korea
- Xenotransplantation Research Center, Seoul National University, Seoul, Korea
| |
Collapse
|
74
|
Dupont PJ, Warrens AN. Fas ligand exerts its pro-inflammatory effects via neutrophil recruitment but not activation. Immunology 2007; 120:133-9. [PMID: 17233740 PMCID: PMC2265864 DOI: 10.1111/j.1365-2567.2006.02504.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Fas ligand (FasL) expression induces apoptosis of activated T cells and has been suggested as a strategy to inhibit graft rejection. Unfortunately, the use of FasL to confer 'immune privilege' in this setting has been hampered by the finding that it may also provoke a destructive granulocytic response. While the Fas/FasL-mediated apoptotic pathways are well defined, the pro-inflammatory effects of FasL are poorly understood. Our aim in this study was to define in vitro the biological effects of FasL on neutrophil recruitment and activation. DAP-3 cells expressing human FasL on the cell membrane (mFasL) potently induced apoptosis in human neutrophils and in activated T lymphocytes. Recombinant human soluble FasL (sFasL), by contrast, was a very weak inducer of apoptosis, even at high concentrations. This latter observation suggests that cleavage of mFasL by naturally occurring matrix metalloproteinases may serve to down-regulate FasL activity in vivo. However, in the presence of a cross-linking antibody, the efficiency of apoptosis-induction by sFasL was greatly increased, suggesting that the lesser pro-apoptotic potency of sFasL reflects an inability to induce trimerization of the Fas receptor. With regard to pro-inflammatory effects, we found that sFasL is a potent neutrophil chemoattractant and, given that it induces little apoptosis, the dominance of sFasL over mFasL may mean that graft-infiltrating neutrophils will survive to mediate inflammation. Neither sFasL nor mFasL produced neutrophil activation as assessed by chemiluminescence assay. This suggests that neutrophils recruited to an inflammatory site by FasL will be activated by mechanisms other than Fas-FasL signalling.
Collapse
Affiliation(s)
- Peter J Dupont
- Department of Immunology, Hammersmith Hospital Campus, Imperial College London, London, United Kingdom
| | | |
Collapse
|
75
|
Abstract
Death receptor ligands (FasL, TRAIL) activate apoptosis in cells expressing the cognate receptors. Evidence suggests that these ligands also deliver pro-inflammatory signals. In the tumor microenvironment, "Fas counterattack" mounted by tumors against immune cells is mediated by tumor-associated FasL. But death ligands crosslinking their receptors also induce inhibition of apoptosis and activation of the transcription factor, NFkappaB, with a subsequent burst of pro-inflammatory cytokine production and tumor growth promotion. NFkappaB, a key link between inflammation and cancer, regulates dual activities of death ligands, depending on molecular signals in the tumor microenvironment. This paper focuses on death ligands as an example of the extensive repertoire of strategies devised by tumors for escape from immune control.
Collapse
Affiliation(s)
- Theresa L Whiteside
- Department of Pathology, Immunology and Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
76
|
Busick RY, Aguilera C, Quinn A. Dominant CTL-inducing epitopes on GAD65 are adjacent to or overlap with dominant Th-inducing epitopes. Clin Immunol 2007; 122:298-311. [PMID: 17174605 DOI: 10.1016/j.clim.2006.10.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2006] [Revised: 10/16/2006] [Accepted: 10/16/2006] [Indexed: 12/22/2022]
Abstract
Immune responses to GAD65 are associated with progression to T1D in NOD mice and humans. Our previous data suggested that dominant CTL-inducing and Th-inducing determinants might preferentially occur in proximal GAD65 sequences. Using a panel of 192 GAD65 peptides we discovered that four of the eight CTL-inducing peptides, including those most biologically relevant, were proximal to previously described I-A(g7)-restricted determinants that characterize natural islet autoimmunity in NOD mice. The CTL determinants 546-554 and 88-98 were presented by GAD65-expressing cells and were displayed on pancreatic LNC, along with 268-278, following beta cell damage. p546-554-specific CTL were detectable in young naive mice and transferred significant islet inflammation into NOD.scid mice. These findings demonstrate that unique regions of GAD65 may be favored during antigen processing, such that diverse dominant epitopes are produced from overlapping sequences, which can engage distinct T cell subsets. Additionally, cross-presentation may enhance GAD65-specific CTL responses in T1D.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Cells, Cultured
- Cytotoxicity, Immunologic
- Diabetes Mellitus, Type 1/enzymology
- Diabetes Mellitus, Type 1/immunology
- Epitopes, T-Lymphocyte/analysis
- Epitopes, T-Lymphocyte/immunology
- Glutamate Decarboxylase/genetics
- Glutamate Decarboxylase/immunology
- H-2 Antigens/immunology
- H-2 Antigens/metabolism
- Immunodominant Epitopes/analysis
- Immunodominant Epitopes/immunology
- Isoenzymes/genetics
- Isoenzymes/immunology
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Molecular Sequence Data
- Peptide Mapping
- T-Lymphocytes, Cytotoxic/enzymology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
Collapse
Affiliation(s)
- Rhea Y Busick
- Department of Biological Sciences, University of Toledo, 2801 W. Bancroft, Toledo, OH 43606, USA
| | | | | |
Collapse
|
77
|
Genestine M, Robert B, Lallemand Y. High-resolution mapping of the Gli3 deletion in the mouse extra-toesH mutant. Genesis 2007; 45:107-12. [PMID: 17304534 DOI: 10.1002/dvg.20270] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Extra-toes is a semidominant mutation that affects the Gli3 gene and provokes limb and brain abnormalities. Among the different alleles of this mutation, Xt(H) is due to a deletion that has not yet been fully characterized. Using a PCR-based strategy, we undertook a high-resolution mapping of this deletion and confirmed that Xt(H) is a null allele of Gli3. We further designed a PCR test to identify unequivocally heterozygous and homozygous embryos from their wild-type littermates. Despite the length of the Xt(H) deletion, available data on the mouse genome indicate that no genes other than Gli3 are deleted in Xt(H) mutants. Thus, the Xt(H) mutation can be used as a model for studying the effects that absence of Gli3 function has during development.
Collapse
|
78
|
Wada A, Tada Y, Kawamura K, Takiguchi Y, Tatsumi K, Kuriyama T, Takenouchi T, O-Wang J, Tagawa M. The effects of FasL on inflammation and tumor survival are dependent on its expression levels. Cancer Gene Ther 2006; 14:262-7. [PMID: 17053813 DOI: 10.1038/sj.cgt.7701008] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The apoptosis-inducing Fas ligand (FasL) is expressed in a variety of human cancers and has been implicated in tumor immune evasion. Paradoxically, ectopic expression of FasL in experimental tumors triggers a neutrophil-mediated inflammatory response and tumor rejection. To resolve these conflicting findings, we have established B16 melanoma and P29 Lewis lung carcinoma lines expressing different levels of FasL and examined their tumorigenicity in vivo. While tumors with a high level of FasL were rapidly rejected as previously reported, those expressing a low level of FasL were not rejected but grew faster than did FasL-negative parental cells. The growth enhancement of FasL(low) tumors was not observed in T-cell-deficient nude mice, suggesting that FasL expressed in tumors at low levels counteracted against T-cell-dependent antitumor responses. In support of this notion, FasL(low) tumors were found to grow faster than parental cells in mice that had acquired tumor-specific immunity. Furthermore, histological examinations revealed apoptosis of lymphocytes in tissue sections of FasL(low) tumors. These results collectively suggest that FasL on tumors is a double-edged sword: at high levels it triggers tumor rejection whereas at low levels it facilitates tumor growth possibly by suppressing antitumor immune responses.
Collapse
Affiliation(s)
- A Wada
- Division of Pathology, Chiba Cancer Center Research Institute, Chiba, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Adachi K, Fujino M, Kitazawa Y, Funeshima-Fuji N, Takahara S, Kimura H, Li XK. Exogenous Expression of Fas-Ligand or CrmA Prolongs the Survival in Rat Liver Transplantation. Transplant Proc 2006; 38:2710-3. [PMID: 17098047 DOI: 10.1016/j.transproceed.2006.08.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Modulation of donor organs by transfection of a gene encoding immmunosuppresive molecules has been recognized as a less toxic approach to prevent allograft rejection. Fas-ligand (FasL) plays a critical role in activation-induced cell death of activated cytotoxic lymphocytes. This may provide a potential for induction of "immune privileged sites" to escape the host immune surveillance system. Cytokine response modifier A (CrmA), a gene product of cowpox virus, blocks caspase as well as perforin/granzyme-mediated apoptotic pathways. Therefore, it may suppress intragraft apoptosis. The aim of the present study was to investigate whether transfection of FasL or CrmA genes prolonged the survival of rat liver allografts. Using the high responder rat combination of DA (RT-1(a)) donor to LEW (RT-1(1)) recipient, we performed orthotopic liver transplantation with subsequent delivery of adenoviral vectors containing FasL, CrmA, or LacZ, at a dose of 1 x 10(9) pfu via a recipient tail vein using a Cre-mediated gene expression system. Recipient survival was assessed as well as immunohistochemical examination of the grafts for anti-CD2, TUNEL, and H&E staining. Statistical analysis was performed with the Mann-Whitney U test. The therapeutic groups showed significantly prolonged recipient survival compared with the LacZ-treated control group. Histologic analysis revealed reduced hepatocyte apoptosis in the CrmA-treated group and increased apoptosis of infiltrating mononuclear cells in the FasL-treated group. These data suggested that FasL and CrmA may be potent genes to prolong rat liver allograft survival.
Collapse
Affiliation(s)
- K Adachi
- Laboratory of Transplantation Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
80
|
Abstract
Immune privilege is a term applied to organs that have a unique relationship with the immune response. These sites prohibit the spread of inflammation, since even minor episodes can threaten organ integrity and function. Once thought to be a passive process relying on physical barriers, immune privilege is now viewed as an active process, which uses multiple mechanisms to maintain organ function. The prototypic organ of immune privilege has been the eye, where the spread of inflammation can threaten vision. Nearly 10 years ago, we discussed the finding that Fas ligand (FasL) was constitutively expressed in the eye and played a major role in immune privilege by inducing apoptosis in inflammatory cells that enter the eye. In this review, we reexamine the original evidence for the role of FasL in immune privilege, update progress on some of the concepts, and discuss some of the issues that remain unresolved.
Collapse
Affiliation(s)
- Thomas A Ferguson
- Department of Ophthalmology and Visual Science, Washington University School of Medicine, St Louis, MO 63110, USA.
| | | |
Collapse
|
81
|
Strauss G, Osen W, Knape I, Jacobsen EM, Müller SM, Debatin KM. Membrane-bound CD95 ligand expressed on human antigen-presenting cells prevents alloantigen-specific T cell response without impairment of viral and third-party T cell immunity. Cell Death Differ 2006; 14:480-8. [PMID: 16902496 DOI: 10.1038/sj.cdd.4402019] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Genetically modified antigen-presenting cells (APC) represent an attractive strategy for in vitro immunomodulation. In the human system, APC expressing HLA-A1 and a membrane-bound form of CD95L (m-CD95L) were used for selective depletion of HLA-A1-specific T cells. In short-term assays, m-CD95L-expressing APC-induced apoptosis in activated T cells and the constitutive presence of m-CD95L and HLA-A1 expressing APC in long-term T cell cultures prevented the expansion of CD4(+) and CD8(+) HLA-A1-specific T cells and the development of HLA-A1-specific cytotoxicity. However, immunity towards third party, viral and bacterial antigens was maintained and T cells spared from depletion could be induced to develop cytotoxicity towards unrelated antigens. Interestingly, inhibition of HLA-A1-specific T cell response absolutely requires the coexpression of m-CD95L and HLA-A1 antigen on the same APC. Thus, m-CD95L expressing APC might be used in clinical settings to obtain tolerance induction in allogeneic transplantation systems or autoimmune diseases.
Collapse
Affiliation(s)
- G Strauss
- University Children's Hospital, Ulm, Germany
| | | | | | | | | | | |
Collapse
|
82
|
Anterior chamber–associated immune deviation and its impact on corneal allograft survival. Curr Opin Organ Transplant 2006. [DOI: 10.1097/01.mot.0000236697.07092.ac] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
83
|
Moore DJ, Markmann JF, Deng S. Avenues for immunomodulation and graft protection by gene therapy in transplantation. Transpl Int 2006; 19:435-45. [PMID: 16771864 DOI: 10.1111/j.1432-2277.2006.00314.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Organ transplantation represents the only definitive therapy for many causes of end-organ failure. However, the universal success of this therapy is limited by chronic allograft rejection, the side effects of chronic immunosuppressive therapy, and a severe shortage of donor organs. Presently, the success of solid-organ transplantation depends on the continuous administration of toxic and nonspecific immunosuppressive agents, therapies that present risks for opportunistic infection, malignancy, and a variety of agent-specific side effects. To promote the use of transplantation with limited risk of long-term sequelae, three dominant research challenges emerge: (i) elimination of the need for exogenous immunosuppression by immunological tolerance induction; (ii) prevention of chronic rejection/graft dysfunction; and (iii) expansion of available organs for transplantation. Gene therapy may provide significant advances and solutions in each of these areas. Rejection of the graft in the immediate post-transplant period has been attacked through the transfer of immunomodulatory molecules in addition to tolerance inducing approaches. Chronic graft rejection may be similarly addressed through permanent tolerance induction or alternatively through the introduction of molecules to resist chronic graft damage. Genetic manipulation of stem cells may ultimately produce transgenic animals to serve as tissue donors to overcome the limited donor organ supply. This review will highlight ongoing developments in the translation of gene therapy approaches to the challenges inherent in transplantation.
Collapse
Affiliation(s)
- Daniel J Moore
- Department of Pediatrics, Vanderbilt University, Nashville, TN, USA
| | | | | |
Collapse
|
84
|
Tan PH, Tan PL, George AJT, Chan CLH. Gene therapy for transplantation with viral vectors – how much of the promise has been realised? Expert Opin Biol Ther 2006; 6:759-72. [PMID: 16856798 DOI: 10.1517/14712598.6.8.759] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Gene therapy holds promise in preventing the development of many diseases. One of the possible applications is the management of organ transplantation. Over the years, advances in vector development have allowed the clinical progression of this form of therapy to become more attainable. Viral vector technology has proved to be better than non-viral vectors at ferrying therapeutic genes to cells. However, many deficiencies in viral vectors hinder the full realisation of gene-based therapy in transplantation. Here, these deficiencies and their ramifications for the future of viral vector development are fully analysed. The authors propose that the slow progress of gene therapy in transplantation may be related to the deficiencies in viral vectors.
Collapse
Affiliation(s)
- Peng H Tan
- Oxford Transplant Centre, Oxford Radcliffe Hospitals NHS Trust, Oxford University, Churchill Hospital, Old Road, Oxford, OX3 7LJ, UK.
| | | | | | | |
Collapse
|
85
|
Narang AS, Mahato RI. Biological and biomaterial approaches for improved islet transplantation. Pharmacol Rev 2006; 58:194-243. [PMID: 16714486 DOI: 10.1124/pr.58.2.6] [Citation(s) in RCA: 137] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Islet transplantation may be used to treat type I diabetes. Despite tremendous progress in islet isolation, culture, and preservation, the clinical use of this modality of treatment is limited due to post-transplantation challenges to the islets such as the failure to revascularize and immune destruction of the islet graft. In addition, the need for lifelong strong immunosuppressing agents restricts the use of this option to a limited subset of patients, which is further restricted by the unmet need for large numbers of islets. Inadequate islet supply issues are being addressed by regeneration therapy and xenotransplantation. Various strategies are being tried to prevent beta-cell death, including immunoisolation using semipermeable biocompatible polymeric capsules and induction of immune tolerance. Genetic modification of islets promises to complement all these strategies toward the success of islet transplantation. Furthermore, synergistic application of more than one strategy is required for improving the success of islet transplantation. This review will critically address various insights developed in each individual strategy and for multipronged approaches, which will be helpful in achieving better outcomes.
Collapse
Affiliation(s)
- Ajit S Narang
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 26 S. Dunlap St., Feurt Building, Room 413, Memphis, TN 38163, USA
| | | |
Collapse
|
86
|
Celik-Ozenci C, Sahin Z, Ustunel I, Akkoyunlu G, Erdogru T, Korgun ET, Baykara M, Demir R. The Fas system may have a role in male reproduction. Fertil Steril 2006; 85 Suppl 1:1168-78. [PMID: 16616089 DOI: 10.1016/j.fertnstert.2005.08.058] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2004] [Revised: 08/17/2005] [Accepted: 08/17/2005] [Indexed: 10/24/2022]
Abstract
OBJECTIVE To assess what the distributions of Fas system proteins are in normal rat testicular tissue; to assess whether there is a change in these distributions and in expression levels with experimentally-induced varicocele of 9, 11, and 13 weeks; and to assess whether there is a relationship between apoptosis and the Fas system in varicocele-induced rat testis. DESIGN Comparative and controlled study. SETTING University animal care and operation unit. ANIMAL(S) Wistar male rats for experimental and control groups. INTERVENTION(S) The control group underwent sham operation (n = 6). Rats in experimental groups underwent partial ligation of the renal vein to induce an experimental varicocele and then were killed at 9 (n = 6), 11 (n = 6), and 13 (n = 6) weeks after induction of varicocele. MAIN OUTCOME MEASURE(S) Tissues were fixed and processed for paraffin and Araldite embedding, and subsequently immunohistochemistry, terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end-labeling, and transmission electron microscopy were performed. In addition, Western blotting was applied. RESULT(S) In control testis, we detected the expression of FasL in spermatids, interestingly at the progressing stages of acrosome formation and in the heads of the spermatozoa being released to lumen. Varicocele induction revealed a significant down-regulation of this protein, especially 11 weeks after the operation, without altering its distribution. Fas protein was present in cytoplasmic extrusions of the elongated spermatids and evidently in Leydig cells of the interstitial tissue. The expression of Fas protein was diminished after 11 weeks of varicocele induction, both in Leydig cells and in cytoplasmic extrusions. The decrease of Fas was significant in the 13-week-old varicocele group, whereas that of FasL was significant in the 11-week-old varicocele group. Compared with sham-operated animals, a minor increase in the number of apoptotic germ cells in varicocele groups was detected. CONCLUSION(S) Our results exposed other possible important roles of the Fas system in addition to than apoptosis in male reproduction. We suggest that the role of the Fas system needs further investigation both in animal models and in human male infertility.
Collapse
Affiliation(s)
- Ciler Celik-Ozenci
- Department of Histology and Embryology, Akdeniz University, Faculty of Medicine, Antalya, Turkey.
| | | | | | | | | | | | | | | |
Collapse
|
87
|
Pearl-Yafe M, Yolcu ES, Yaniv I, Stein J, Shirwan H, Askenasy N. The dual role of Fas-ligand as an injury effector and defense strategy in diabetes and islet transplantation. Bioessays 2006; 28:211-22. [PMID: 16435302 DOI: 10.1002/bies.20356] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The exact process that leads to the eruption of autoimmune reactions against beta cells and the evolution of diabetes is not fully understood. Macrophages and T cells may launch an initial immune reaction against the pancreatic islets of Langerhans, provoking inflammation and destructive insulitis. The information on the molecular mechanisms of the emergence of beta cell injury is controversial and points to possibly important roles for the perforin-granzyme, Fas-Fas-ligand (FasL) and tumor-necrosis-factor-mediated apoptotic pathways. FasL has several unique features that make it a potentially ideal immunomodulatory tool. Most important, FasL is selectively toxic to cytotoxic T cells and less harmful to regulatory T cells. This review discusses the intrinsic sensitivity of beta cells to FasL-mediated apoptosis, the conditions that underlie this beta cell sensitivity, and the feasibility of using FasL to arrest autoimmunity and prevent islet allograft rejection. In both the autoimmune and transplant settings, it is imperative to progress from the administration of nonspecific immunosuppressive therapy to the concept of beta-cell-specific immunomodulation. FasL evolves as a prime candidate for antigen-specific immunomodulation.
Collapse
Affiliation(s)
- Michal Pearl-Yafe
- Frankel Laboratory of Experimental Bone Marrow Transplantation, Department of Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Israel
| | | | | | | | | | | |
Collapse
|
88
|
Kawamoto K, Tanemura M, Nishida T, Fukuzawa M, Ito T, Matsuda H. Significant Inhibition of Human CD8+ Cytotoxic T Lymphocyte-Mediated Xenocytotoxicity by Overexpression of the Human Decoy Fas Antigen. Transplantation 2006; 81:789-96. [PMID: 16534484 DOI: 10.1097/01.tp.0000199266.07237.25] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Human CD8(+) CTL-mediated killing may be important for xenograft rejection. The purpose of this study was to explore the preventing methods for CTL-mediated xenocytotoxicity by overexpression of human decoy Fas, which lacks a death domain in its cytoplasmic region, by binding competition with endogenous pig Fas. Moreover, the cytoprotective effect of this CTL-killing of membrane-bound human FasL, which is resistant to metalloproteolytic cleavage, was also assessed. METHODS Human CTL were generated by the stimulation of human PBMC with swine endothelial cells (SEC) and human IL-2, subsequently a CD8(+) population were selected by magnetic beads and employed as the effector cells. Stable SEC transfectants expressing either decoy Fas or membrane-bound FasL were established. Double-transfectants were also created. The amelioration of cytotoxicity to these transfectants was examined with Cr release assay. RESULTS.: Human CD8(+) CTL were highly detrimental against parental SEC. This CTL-killing was strongly inhibited by anti-FasL mAb treatment, however partial suppression was observed by Concanamycin A treatment. The overexpression of either decoy Fas or membrane-bound FasL in SEC markedly inhibited CTL-xenocytotoxicity. The double expressions of these molecules also significantly reduced this xenocytotoxicity despite the low levels of expression of either decoy Fas or membrane-bound FasL. CONCLUSION These findings indicate that the strong xenocytotoxicity of human CD8(+) CTL is mediated mainly by the Fas/FasL pathway. The overexpression of either decoy Fas or membrane-bound FasL were quite effective in preventing CTL-killing. Furthermore, the combined expression of both molecules in pig cells may create a window of opportunity for prolonging xenograft survival.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Antigens, Heterophile/immunology
- Cytotoxicity, Immunologic/drug effects
- Fas Ligand Protein
- Graft Rejection/immunology
- Graft Rejection/prevention & control
- Humans
- Membrane Glycoproteins/antagonists & inhibitors
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Membrane Glycoproteins/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor, Member 6b
- Sequence Deletion
- Swine/immunology
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- Transplantation, Heterologous/immunology
- Tumor Necrosis Factor Inhibitors
- Tumor Necrosis Factors/immunology
- Tumor Necrosis Factors/metabolism
- Up-Regulation
Collapse
Affiliation(s)
- Koichi Kawamoto
- Department of Surgery (E1), Osaka University Graduate School of Medicine, Osaka, Japan
| | | | | | | | | | | |
Collapse
|
89
|
Abstract
It has become clear in recent years that apoptosis is a normal process in trophoblast turnover during pregnancy. Increased trophoblast apoptosis has been observed in the placenta of women with preeclampsia, serum from women with preeclampsia has been found to induce increased trophoblast sensitivity to Fas-mediated apoptosis, and serum from women with preeclampsia has elevated levels of various chemokines, growth factors and cytokines that are involved in the regulation of apoptosis. This review highlights the importance of apoptosis in normal placental development and explores the mechanisms whereby Fas-mediated apoptosis may play a role in conditions related to abnormal placentation, such as preeclampsia.
Collapse
Affiliation(s)
- Donna M Neale
- Department of Gynecology and Obstetrics, John Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | |
Collapse
|
90
|
Abstract
The most intensively studied autoimmune disorder, type 1 diabetes mellitus (DM1), has attracted perhaps the greatest interest for gene-based therapeutic and prophylactic interventions. The final clinical manifestation of this immunologically and genetically complex disease, the absence of insulin, is the major starting point for almost all the gene therapy modalities attempted to date. Insulin replacement by transplantation of islets of Langerhans or surrogate beta cells is the obvious choice, but the allogeneic nature of the transplants activates potent antidonor immunoreactivity necessitating gene and cell-based immunosuppressive strategies as an alternative to the toxic pharmacologic immunosuppressives indicated for classic solid organ transplants. Accumulating knowledge of the cellular mechanisms involved in onset, however, have yielded promising tolerance induction prophylactic approaches using genes and cells. Despite the early successes in a number of animal models, the true test of efficacy in humans remains to be demonstrated.
Collapse
Affiliation(s)
- Nick Giannoukakis
- Diabetes Institute, Pediatric Research Section, Children's Hospital of Pittsburgh and University of Pittsburgh, Rangos Research Center, Pittsburgh, PA 15213, USA
| | | |
Collapse
|
91
|
Kotani N, Fukuo K, Yasuda O, Sugimoto K, Katuya T, Takemura Y, Kawamoto H, Yokoi T, Suzuki A, Ogihara T. Fas Ligand mRNA Levels of Circulating Leukocytes Reflect Endothelial Dysfunction in Hyperlipidemic but Not in Non-Hyperlipidemic Patients. Hypertens Res 2006; 29:217-25. [PMID: 16778328 DOI: 10.1291/hypres.29.217] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
To find a novel marker for identifying patients at high-risk for endothelial dysfunction among patients with atherosclerosis, we examined the correlation between mRNA levels of Fas ligand (FasL), an apoptosis-inducing factor, in circulating leukocytes and clinical parameters in these patients. FasL mRNA levels of circulating leukocytes were measured with the TaqMan-PCR method. A negative correlation was observed between brachial artery flow-mediated dilatation (%FMD) and FasL mRNA levels of leukocytes in hyperlipidemic but not in non-hyperlipidemic patients. %FMD was more impaired in patients with a high level of FasL mRNA than in those with a low level of FasL mRNA. Interestingly, the improvement of %FMD by treatment with a 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor (simvastatin) was greater in the group showing a decrease in FasL mRNA than in the group with no such decrease. Additionally, simvastatin suppressed the FasL mRNA expression in leukocytes and decreased plasma oxidized low-density lipoprotein (OxLDL) levels. Furthermore, the supernatant of cultured leukocytes from hyperlipidemic patients induced cell death in Jurkat T cells, which was neutralized by an antibody against FasL. These findings suggest that high FasL mRNA expression in circulating leukocytes may be a marker of high-risk for endothelial dysfunction in hyperlipidemic but not in non-hyperlipidemic patients. This information may provide a novel basis for targeting of statin therapy in patients with vulnerable plaques.
Collapse
Affiliation(s)
- Noriko Kotani
- Department of Geriatric Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
92
|
Kidoya H, Umemura M, Kawabe T, Matsuzaki G, Yahagi A, Imamura R, Suda T. Fas Ligand Induces Cell-Autonomous IL-23 Production in Dendritic Cells, a Mechanism for Fas Ligand-Induced IL-17 Production. THE JOURNAL OF IMMUNOLOGY 2005; 175:8024-31. [PMID: 16339539 DOI: 10.4049/jimmunol.175.12.8024] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Fas ligand (FasL) has the potential to induce inflammation accompanied by massive neutrophil infiltration. We previously reported that FasL rapidly induces the production of various inflammatory cytokines including IL-1beta and IL-17. In this study, we investigated the mechanism of the FasL-induced IL-17 production. We found that the culture supernatant of mouse resident peritoneal exudate cells (PEC) cocultured with FasL-expressing tumor (FFL) cells induced IL-17 production in freshly isolated resident PEC. Anti-IL-1beta Ab strongly inhibited the IL-17-inducing activity. However, rIL-1beta by itself induced only weak IL-17 production. Intriguingly, anti-IL-12 Ab but not an IL-15-neutralizing agent, IL15R-Fc, strongly inhibited the FasL-induced IL-17-inducing activity. IL-23, which shares the p40 subunit with IL-12, but not IL-12 itself, induced IL-17 production synergistically with IL-1beta in resident PEC. FasL induced the production of IL-23 in PEC in vivo and in vitro, and IL-17 production following the i.p. injection of FFL cells was severely impaired in p40-/- mice, indicating that IL-23 plays an important role in the FasL-induced IL-17 production. FFL also induced the production of IL-23 in bone marrow- or PEC-derived dendritic cells (DCs). Finally, FasL induced only weak p40 production in a mixture of p40-/- and Fas-/- DC, indicating that FasL induces IL-23 production in DC mainly in a cell-autonomous manner.
Collapse
Affiliation(s)
- Hiroyasu Kidoya
- Center for the Development of Molecular Target Drugs, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | | | | | | | | | | | | |
Collapse
|
93
|
Straszewski-Chavez SL, Abrahams VM, Mor G. The role of apoptosis in the regulation of trophoblast survival and differentiation during pregnancy. Endocr Rev 2005; 26:877-97. [PMID: 15901666 DOI: 10.1210/er.2005-0003] [Citation(s) in RCA: 206] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Apoptosis is important for normal placental development, but it may also be involved in the pathophysiology of pregnancy-related diseases. Normal placental development is dependent upon the differentiation and invasion of the trophoblast, the main cellular component of the placenta. Trophoblast apoptosis increases in normal placentas as gestation proceeds, and a greater incidence of trophoblast apoptosis has been observed in pregnancies complicated by preeclampsia or intrauterine growth retardation (IUGR). In response to different stimuli, apoptosis may be initiated extrinsically by the death receptor pathway or intrinsically by the mitochondrial pathway. The central executioners of apoptosis are the caspases, which cleave numerous vital cellular proteins to affect the apoptotic cascade. By inhibiting caspase activation, several endogenous inhibitors, including flice-like inhibitory proteins (FLIPs), inhibitors of apoptosis (IAPs), and antiapoptotic Bcl-2 family members, can prevent further propagation of the death signal. Macrophages present at the maternal-fetal interface may also contribute to trophoblast survival by removing apoptotic cells and producing cytokines and growth factors, which influence the progression of the apoptotic cascade. This review focuses on the role of apoptosis in trophoblast development and differentiation, the molecular mechanisms by which normal trophoblast apoptosis can occur, and how it is regulated to prevent excessive trophoblast apoptosis and possible pregnancy complications.
Collapse
Affiliation(s)
- Shawn L Straszewski-Chavez
- Department of Obstetrics, Gynecology and Reproductive Sciences, Reproductive Immunology Unit, Yale University School of Medicine, 333 Cedar Street FMB 301, New Haven, CT 06520, USA
| | | | | |
Collapse
|
94
|
Matute-Bello G, Lee JS, Liles WC, Frevert CW, Mongovin S, Wong V, Ballman K, Sutlief S, Martin TR. Fas-mediated acute lung injury requires fas expression on nonmyeloid cells of the lung. THE JOURNAL OF IMMUNOLOGY 2005; 175:4069-75. [PMID: 16148156 DOI: 10.4049/jimmunol.175.6.4069] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Fas (CD95) is a membrane surface receptor, which, in the lungs, is expressed in macrophages, neutrophils, and epithelial cells. In mice, Fas activation leads to a form of lung injury characterized by increased alveolar permeability. We investigated whether Fas-mediated lung injury occurs primarily as a result of Fas activation in myeloid cells (such as macrophages) or in nonmyeloid cells (such as epithelial cells). Chimeric mice lacking Fas in either myeloid or nonmyeloid cells were generated by transplanting marrow cells from lpr mice (which lack Fas) into lethally irradiated C57BL/6 mice (MyFas(-) group) or vice versa (MyFas(+) group). Additional mice transplanted with marrow cells from their same strain served as controls (Fas(+) ctr and Fas(-) ctr groups). Sixty days after transplantation, the mice received intratracheal instillations of the Fas-activating mAb Jo2 (n = 10/group), or an isotype control Ab (n = 10/group), and were euthanized 24-h later. Only animals expressing Fas in nonmyeloid cells (Fas(+) ctr and MyFas(-)) showed significant increases in lung neutrophil content and in alveolar permeability. These same mice showed tissue evidence of lung injury and caspase-3 activation in cells of the alveolar walls. Despite differences in the neutrophilic response and lung injury, there was no statistical difference in the lung cytokine concentrations (KC and MIP-2) among groups. We conclude that Fas-mediated lung injury requires expression of Fas on nonmyeloid cells of the lungs. These findings suggest that the alveolar epithelium is the primary target of Fas-mediated acute lung injury, and demonstrate that apoptotic processes may be associated with neutrophilic inflammation.
Collapse
Affiliation(s)
- Gustavo Matute-Bello
- Division of Pulmonary and Critical Care Medicine, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Sun J, Hilliard B, Xu L, Chen YH. Essential roles of the Fas-associated death domain in autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2005; 175:4783-8. [PMID: 16177127 DOI: 10.4049/jimmunol.175.7.4783] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The Fas-associated death domain (FADD) protein mediates apoptosis by coupling death receptors with the caspase cascade. Paradoxically, it also promotes cell mitosis through its C-terminal region. Apoptosis and mitosis are opposing processes that can have radically different consequences. To determine which of the FADD effects prevails in T cell-mediated autoimmune diseases, we studied myelin oligodendrocyte glycoprotein-induced experimental autoimmune encephalomyelitis (EAE) using mice that express a dominant-negative FADD (FADD-DN) transgene in the T cell lineage. We found that FADD blockade in T cells prevented the development of autoimmune encephalomyelitis and inhibited both Th1 and Th2 type responses. Myelin oligodendrocyte glycoprotein-specific T cell proliferation was also dramatically reduced in FADD-DN mice despite the resistance of T cells to activation-induced cell death. These results indicate that although FADD expressed by T cells is involved in regulating both mitosis and apoptosis, its effect on mitosis prevails in EAE, and that strategies inhibiting FADD functions in T cells could be effective in preventing the disease.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/antagonists & inhibitors
- Adaptor Proteins, Signal Transducing/genetics
- Adaptor Proteins, Signal Transducing/physiology
- Animals
- Apoptosis/immunology
- Cell Differentiation/immunology
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Fas-Associated Death Domain Protein
- Lymphocyte Activation/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Mitosis
- Protein Structure, Tertiary
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Jing Sun
- Department of Pathology and Laboratory Medicine, School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
96
|
Tan PH, Chan CLH, Chan C, George AJT. The evolving role of gene-based treatment in surgery. Br J Surg 2005; 92:1466-80. [PMID: 16273530 DOI: 10.1002/bjs.5181] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Abstract
Background
The completion of the sequencing of the human genome in 2003 marked the dawn of a new era of human biology and medicine. Although these remarkable scientific advances improve the understanding of human biology, the question remains how this rapidly expanding knowledge of functional genomics affects the role of surgeons. This article reviews the potential therapeutic application of gene therapy for various surgical conditions.
Methods
The core of this review was derived from a Medline database literature search.
Results and conclusion
The currently available vectors in the field of gene therapy and their limitations for clinical applications were analysed. The achievements of gene therapy in clinical trials and the future ramifications for surgery were also explored. Whether gene therapy takes a major role in surgical practice will depend greatly on the success of future vector development. Advances in viral vector technology to reduce the inflammatory effect, and improvements in the efficiency of gene delivery using non-viral vector technology, would allow this form of therapy to become more clinically applicable.
Collapse
Affiliation(s)
- P H Tan
- Department of Surgery, Stoke Mandeville Hospital, South Buckinghamshire NHS Trust, Aylesbury, UK.
| | | | | | | |
Collapse
|
97
|
Ryan AE, Shanahan F, O'Connell J, Houston AM. Addressing the "Fas counterattack" controversy: blocking fas ligand expression suppresses tumor immune evasion of colon cancer in vivo. Cancer Res 2005; 65:9817-23. [PMID: 16267003 DOI: 10.1158/0008-5472.can-05-1462] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Fas ligand (FasL/CD95L) is a transmembrane protein belonging to the tumor necrosis factor superfamily that can trigger apoptotic cell death following ligation to its receptor, Fas (CD95/APO-1). Expression of FasL may help to maintain tumor cells in a state of immune privilege by inducing apoptosis of antitumor immune effector cells-the "Fas counterattack." However, the ability of FasL to mediate tumor immune privilege is controversial due to studies that indicate FasL has both pro- and anti-inflammatory activities. To resolve this controversy and functionally define the role of FasL in tumor immune evasion, we investigated if suppression of endogenously expressed FasL in colon tumor cells resulted in reduced tumor development and improved antitumor immune challenge in vivo. Specifically, FasL expression in CMT93 colon carcinoma cells was down-regulated following stable transfection with a plasmid encoding antisense FasL cDNA. Down-regulation of FasL expression had no effect on tumor growth in vitro but significantly reduced tumor development in syngeneic immunocompetent mice in vivo. Tumor size was also significantly decreased. Reduced FasL expression by tumor cells led to increased lymphocyte infiltration. The overall level of neutrophils present in all of the tumors examined was low, with no difference between the tumors, irrespective of FasL expression. Thus, down-regulation of FasL expression by colon tumor cells results in an improved antitumor immune challenge in vivo, providing functional evidence in favor of the "Fas counterattack" as a mechanism of tumor immune evasion.
Collapse
Affiliation(s)
- Aideen E Ryan
- Department of Medicine, Cork University Hospital, and Alimentary Pharmabiotic Centre, National University of Ireland Cork, Cork, Ireland
| | | | | | | |
Collapse
|
98
|
Igney FH, Krammer PH. Tumor counterattack: fact or fiction? Cancer Immunol Immunother 2005; 54:1127-36. [PMID: 15889255 PMCID: PMC11034178 DOI: 10.1007/s00262-005-0680-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2004] [Accepted: 01/24/2005] [Indexed: 12/30/2022]
Abstract
Cancer development relies on a variety of mechanisms that facilitate tumor growth despite the presence of a functioning immune system. Understanding these mechanisms may foster novel therapeutic approaches for oncology and organ transplantation. By expression of the apoptosis-inducing protein CD95L (FasL, APO-1L, CD178), tumors may eliminate tumor-infiltrating lymphocytes and suppress anti-tumor immune responses, a phenomenon called "tumor counterattack". On the one hand, preliminary evidence of tumor counterattack in human tumors exists, and CD95L expression can prevent T-cell responses in vitro. On the other hand, CD95L-expressing tumors are rapidly rejected and induce inflammation in mice. Here, we summarize and discuss the consequences of CD95L expression of tumor cells and its contribution to immune escape.
Collapse
Affiliation(s)
- Frederik H Igney
- Tumor Immunology Program, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | | |
Collapse
|
99
|
Park SM, Schickel R, Peter ME. Nonapoptotic functions of FADD-binding death receptors and their signaling molecules. Curr Opin Cell Biol 2005; 17:610-6. [PMID: 16226446 DOI: 10.1016/j.ceb.2005.09.010] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2005] [Accepted: 09/29/2005] [Indexed: 02/01/2023]
Abstract
Death receptors (DRs) are surface receptors that when triggered have the capacity to induce apoptosis in cells by forming the death-inducing signaling complex (DISC). The first protein recruited to form the DISC is the adaptor protein FADD/Mort1. Some members of the DR family, CD95 and the TRAIL receptors DR4 and DR5, directly bind FADD, whereas others, such as TNF receptor I and DR3, initially bind another adaptor protein, TRADD, which then recruits FADD. While all DRs can activate both apoptotic and non-apoptotic pathways, it has been widely assumed that the main physiological role of FADD-binding death receptors is to trigger apoptosis. However, recent work has ascribed multiple non-apoptotic activities to these receptors and/or the signaling components of the DISC.
Collapse
Affiliation(s)
- Sun-Mi Park
- The Ben May Institute for Cancer Research, University of Chicago, 924 E. 57th Street., Chicago, Illinois 60637, USA
| | | | | |
Collapse
|
100
|
O'Brien DI, Nally K, Kelly RG, O'Connor TM, Shanahan F, O'Connell J. Targeting the Fas/Fas ligand pathway in cancer. Expert Opin Ther Targets 2005; 9:1031-44. [PMID: 16185156 DOI: 10.1517/14728222.9.5.1031] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Fas is a transmembrane receptor that can induce apoptosis after cross-linking with either agonistic antibodies or with Fas ligand (FasL). Although originally described as an important regulator of peripheral immune homeostasis, accumulating evidence suggests that the Fas/FasL system plays an important role in tumour development. In addition to its proapoptotic functions, accumulating evidence demonstrates that Fas can activate numerous nonapoptotic signalling pathways, and that activation of these pathways can result in increased tumourigenicity and metastasis. This review summarises the current understanding of the Fas/FasL system in tumorigenesis and discusses attempts to utilise the Fas/FasL system in the treatment of cancer.
Collapse
Affiliation(s)
- Darren I O'Brien
- Department of Medicine, NUIC, Clinical Sciences Building, University Hospital, Cork, Ireland
| | | | | | | | | | | |
Collapse
|