1
|
Use of the Human Granulysin Transgenic Mice To Evaluate the Role of Granulysin Expression by CD8 T Cells in Immunity To Mycobacterium tuberculosis. mBio 2022; 13:e0302022. [PMID: 36409085 PMCID: PMC9765553 DOI: 10.1128/mbio.03020-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The cytotoxic granules of human NK and CD8 T cells contain the effector molecule granulysin. Although in vitro studies indicate that granulysin is bactericidal to Mycobacterium tuberculosis and human CD8 T cells restrict intracellular M. tuberculosis by granule exocytosis, the role of granulysin in cell-mediated immunity against infection is incompletely understood, in part because a granulysin gene ortholog is absent in mice. Transgenic mice that express human granulysin (GNLY-Tg) under the control of human regulatory DNA sequences permit the study of granulysin in vivo. We assessed whether granulysin expression by murine CD8 T cells enhances their control of M. tuberculosis infection. GNLY-Tg mice did not control pulmonary M. tuberculosis infection better than non-Tg control mice, and purified GNLY-Tg and non-Tg CD8 T cells had a similar ability to transfer protection to T cell deficient mice. Lung CD8 T cells from infected control and GNLY-transgenic mice similarly controlled intracellular M. tuberculosis growth in macrophages in vitro. Importantly, after M. tuberculosis infection of GNLY-Tg mice, granulysin was detected in NK cells but not in CD8 T cells. Only after prolonged in vitro stimulation could granulysin expression be detected in antigen-specific CD8 T cells. GNLY-Tg mice are an imperfect model to determine whether granulysin expression by CD8 T cells enhances immunity against M. tuberculosis. Better models expressing granulysin are needed to explore the role of this antimicrobial effector molecule in vivo. IMPORTANCE Human CD8 T cells express the antimicrobial peptide granulysin in their cytotoxic granules, and in vitro analysis suggest that it restricts growth of Mycobacterium tuberculosis and other intracellular pathogens. The murine model of tuberculosis cannot assess granulysin's role in vivo, as rodents lack the granulysin gene. A long-held hypothesis is that murine CD8 T cells inefficiently control M. tuberculosis infection because they lack granulysin. We used human granulysin transgenic (GNLY-Tg) mice to test this hypothesis. GNLY-Tg mice did not differ in their susceptibility to tuberculosis. However, granulysin expression by pulmonary CD8 T cells could not be detected after M. tuberculosis infection. As the pattern of granulysin expression in human CD8 T cells and GNLY-Tg mice seem to differ, GNLY-Tg mice are an imperfect model to study the role of granulysin. An improved model is needed to answer the importance of granulysin expression by CD8 T cells in different diseases.
Collapse
|
2
|
Chan Y, Fong S, Poh C, Carissimo G, Yeo NK, Amrun SN, Goh YS, Lim J, Xu W, Chee RS, Torres‐Ruesta A, Lee CY, Tay MZ, Chang ZW, Lee W, Wang B, Tan S, Kalimuddin S, Young BE, Leo Y, Wang C, Lee B, Rötzschke O, Lye DC, Renia L, Ng LFP. Asymptomatic COVID-19: disease tolerance with efficient anti-viral immunity against SARS-CoV-2. EMBO Mol Med 2021; 13:e14045. [PMID: 33961735 PMCID: PMC8185544 DOI: 10.15252/emmm.202114045] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 01/08/2023] Open
Abstract
The immune responses and mechanisms limiting symptom progression in asymptomatic cases of SARS-CoV-2 infection remain unclear. We comprehensively characterized transcriptomic profiles, cytokine responses, neutralization capacity of antibodies, and cellular immune phenotypes of asymptomatic patients with acute SARS-CoV-2 infection to identify potential protective mechanisms. Compared to symptomatic patients, asymptomatic patients had higher counts of mature neutrophils and lower proportion of CD169+ expressing monocytes in the peripheral blood. Systemic levels of pro-inflammatory cytokines were also lower in asymptomatic patients, accompanied by milder pro-inflammatory gene signatures. Mechanistically, a more robust systemic Th2 cell signature with a higher level of virus-specific Th17 cells and a weaker yet sufficient neutralizing antibody profile against SARS-CoV-2 was observed in asymptomatic patients. In addition, asymptomatic COVID-19 patients had higher systemic levels of growth factors that are associated with cellular repair. Together, the data suggest that asymptomatic patients mount less pro-inflammatory and more protective immune responses against SARS-CoV-2 indicative of disease tolerance. Insights from this study highlight key immune pathways that could serve as therapeutic targets to prevent disease progression in COVID-19.
Collapse
Affiliation(s)
- Yi‐Hao Chan
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and ResearchSingapore CitySingapore
- Singapore Immunology Network, Agency for Science, Technology and ResearchSingapore CitySingapore
| | - Siew‐Wai Fong
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and ResearchSingapore CitySingapore
- Singapore Immunology Network, Agency for Science, Technology and ResearchSingapore CitySingapore
| | - Chek‐Meng Poh
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and ResearchSingapore CitySingapore
- Singapore Immunology Network, Agency for Science, Technology and ResearchSingapore CitySingapore
| | - Guillaume Carissimo
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and ResearchSingapore CitySingapore
- Singapore Immunology Network, Agency for Science, Technology and ResearchSingapore CitySingapore
| | - Nicholas Kim‐Wah Yeo
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and ResearchSingapore CitySingapore
- Singapore Immunology Network, Agency for Science, Technology and ResearchSingapore CitySingapore
| | - Siti Naqiah Amrun
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and ResearchSingapore CitySingapore
- Singapore Immunology Network, Agency for Science, Technology and ResearchSingapore CitySingapore
| | - Yun Shan Goh
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and ResearchSingapore CitySingapore
- Singapore Immunology Network, Agency for Science, Technology and ResearchSingapore CitySingapore
| | - Jackwee Lim
- Singapore Immunology Network, Agency for Science, Technology and ResearchSingapore CitySingapore
| | - Weili Xu
- Singapore Immunology Network, Agency for Science, Technology and ResearchSingapore CitySingapore
| | - Rhonda Sin‐Ling Chee
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and ResearchSingapore CitySingapore
- Singapore Immunology Network, Agency for Science, Technology and ResearchSingapore CitySingapore
| | - Anthony Torres‐Ruesta
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and ResearchSingapore CitySingapore
- Singapore Immunology Network, Agency for Science, Technology and ResearchSingapore CitySingapore
- Department of BiochemistryYong Loo Lin School of MedicineNational University of SingaporeSingapore CitySingapore
| | - Cheryl Yi‐Pin Lee
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and ResearchSingapore CitySingapore
- Singapore Immunology Network, Agency for Science, Technology and ResearchSingapore CitySingapore
| | - Matthew Zirui Tay
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and ResearchSingapore CitySingapore
- Singapore Immunology Network, Agency for Science, Technology and ResearchSingapore CitySingapore
| | - Zi Wei Chang
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and ResearchSingapore CitySingapore
- Singapore Immunology Network, Agency for Science, Technology and ResearchSingapore CitySingapore
| | - Wen‐Hsin Lee
- Singapore Immunology Network, Agency for Science, Technology and ResearchSingapore CitySingapore
| | - Bei Wang
- Singapore Immunology Network, Agency for Science, Technology and ResearchSingapore CitySingapore
| | - Seow‐Yen Tan
- Department of Infectious DiseasesChangi General HospitalSingapore CitySingapore
| | - Shirin Kalimuddin
- Department of Infectious DiseasesSingapore General HospitalSingapore CitySingapore
- Emerging Infectious Diseases ProgramDuke‐NUS Medical SchoolSingapore CitySingapore
| | - Barnaby Edward Young
- National Centre for Infectious DiseasesSingapore CitySingapore
- Department of Infectious DiseasesTan Tock Seng HospitalSingapore CitySingapore
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingapore CitySingapore
| | - Yee‐Sin Leo
- National Centre for Infectious DiseasesSingapore CitySingapore
- Department of Infectious DiseasesTan Tock Seng HospitalSingapore CitySingapore
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingapore CitySingapore
- Saw Swee Hock School of Public HealthNational University of Singapore and National University Health SystemSingapore CitySingapore
- Yong Loo Lin School of MedicineNational University of SingaporeSingapore CitySingapore
| | - Cheng‐I Wang
- Singapore Immunology Network, Agency for Science, Technology and ResearchSingapore CitySingapore
| | - Bernett Lee
- Singapore Immunology Network, Agency for Science, Technology and ResearchSingapore CitySingapore
| | - Olaf Rötzschke
- Singapore Immunology Network, Agency for Science, Technology and ResearchSingapore CitySingapore
| | - David Chien Lye
- National Centre for Infectious DiseasesSingapore CitySingapore
- Department of Infectious DiseasesTan Tock Seng HospitalSingapore CitySingapore
- Lee Kong Chian School of MedicineNanyang Technological UniversitySingapore CitySingapore
- Yong Loo Lin School of MedicineNational University of SingaporeSingapore CitySingapore
| | - Laurent Renia
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and ResearchSingapore CitySingapore
- Singapore Immunology Network, Agency for Science, Technology and ResearchSingapore CitySingapore
| | - Lisa F P Ng
- A*STAR Infectious Diseases Labs (A*STAR ID Labs)Agency for Science, Technology and ResearchSingapore CitySingapore
- Singapore Immunology Network, Agency for Science, Technology and ResearchSingapore CitySingapore
- Department of BiochemistryYong Loo Lin School of MedicineNational University of SingaporeSingapore CitySingapore
- Institute of Infection, Veterinary and Ecological SciencesUniversity of LiverpoolLiverpool, LiverpoolUK
| |
Collapse
|
3
|
Afshar-Sterle S, Zotos D, Bernard NJ, Scherger AK, Rödling L, Alsop AE, Walker J, Masson F, Belz GT, Corcoran LM, O'Reilly LA, Strasser A, Smyth MJ, Johnstone R, Tarlinton DM, Nutt SL, Kallies A. Fas ligand-mediated immune surveillance by T cells is essential for the control of spontaneous B cell lymphomas. Nat Med 2014; 20:283-90. [PMID: 24487434 DOI: 10.1038/nm.3442] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 12/04/2013] [Indexed: 12/23/2022]
Abstract
Loss of function of the tumor suppressor gene PRDM1 (also known as BLIMP1) or deregulated expression of the oncogene BCL6 occurs in a large proportion of diffuse large B cell lymphoma (DLBCL) cases. However, targeted mutation of either gene in mice leads to only slow and infrequent development of malignant lymphoma, and despite frequent mutation of BCL6 in activated B cells of healthy individuals, lymphoma development is rare. Here we show that T cells prevent the development of overt lymphoma in mice caused by Blimp1 deficiency or overexpression of Bcl6 in the B cell lineage. Impairment of T cell control results in rapid development of DLBCL-like disease, which can be eradicated by polyclonal CD8(+) T cells in a T cell receptor-, CD28- and Fas ligand-dependent manner. Thus, malignant transformation of mature B cells requires mutations that impair intrinsic differentiation processes and permit escape from T cell-mediated tumor surveillance.
Collapse
Affiliation(s)
- Shoukat Afshar-Sterle
- 1] Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia. [2] Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia. [3]
| | - Dimitra Zotos
- 1] Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia. [2] Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia. [3]
| | - Nicholas J Bernard
- 1] Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia. [2] Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia. [3]
| | - Anna K Scherger
- 1] Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia. [2] Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Lisa Rödling
- 1] Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia. [2] Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Amber E Alsop
- 1] Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia. [2] Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Jennifer Walker
- 1] Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia. [2] Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Frederick Masson
- 1] Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia. [2] Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Gabrielle T Belz
- 1] Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia. [2] Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Lynn M Corcoran
- 1] Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia. [2] Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Lorraine A O'Reilly
- 1] Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia. [2] Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Andreas Strasser
- 1] Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia. [2] Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Mark J Smyth
- 1] Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia. [2] School of Medicine, University of Queensland, Herston, Queensland, Australia
| | - Ricky Johnstone
- 1] Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia. [2] Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - David M Tarlinton
- 1] Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia. [2] Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Stephen L Nutt
- 1] Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia. [2] Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Axel Kallies
- 1] Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia. [2] Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
4
|
Wolchinsky R, Hod-Marco M, Oved K, Shen-Orr SS, Bendall SC, Nolan GP, Reiter Y. Antigen-dependent integration of opposing proximal TCR-signaling cascades determines the functional fate of T lymphocytes. THE JOURNAL OF IMMUNOLOGY 2014; 192:2109-19. [PMID: 24489091 DOI: 10.4049/jimmunol.1301142] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
T cell anergy is a key tolerance mechanism to mitigate unwanted T cell activation against self by rendering lymphocytes functionally inactive following Ag encounter. Ag plays an important role in anergy induction where high supraoptimal doses lead to the unresponsive phenotype. How T cells "measure" Ag dose and how this determines functional output to a given antigenic dose remain unclear. Using multiparametric phospho-flow and mass cytometry, we measured the intracellular phosphorylation-dependent signaling events at a single-cell resolution and studied the phosphorylation levels of key proximal human TCR activation- and inhibition-signaling molecules. We show that the intracellular balance and signal integration between these opposing signaling cascades serve as the molecular switch gauging Ag dose. An Ag density of 100 peptide-MHC complexes/cell was found to be the transition point between dominant activation and inhibition cascades, whereas higher Ag doses induced an anergic functional state. Finally, the neutralization of key inhibitory molecules reversed T cell unresponsiveness and enabled maximal T cell functions, even in the presence of very high Ag doses. This mechanism permits T cells to make integrated "measurements" of Ag dose that determine subsequent functional outcomes.
Collapse
Affiliation(s)
- Ron Wolchinsky
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | | | | | | | | | | | | |
Collapse
|
5
|
Davis LC, Morgan AJ, Chen JL, Snead CM, Bloor-Young D, Shenderov E, Stanton-Humphreys MN, Conway SJ, Churchill GC, Parrington J, Cerundolo V, Galione A. NAADP activates two-pore channels on T cell cytolytic granules to stimulate exocytosis and killing. Curr Biol 2012. [PMID: 23177477 PMCID: PMC3525857 DOI: 10.1016/j.cub.2012.10.035] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
A cytotoxic T lymphocyte (CTL) kills an infected or tumorigenic cell by Ca2+-dependent exocytosis of cytolytic granules at the immunological synapse formed between the two cells. Although inositol 1,4,5-trisphosphate (IP3)-mediated Ca2+ release from the endoplasmic reticulum activates the store-operated Ca2+-influx pathway that is necessary for exocytosis, it is not a sufficient stimulus [1–4]. Here we identify the Ca2+-mobilizing messenger nicotinic acid adenine dinucleotide phosphate (NAADP) and its recently identified molecular target, two-pore channels (TPCs) [5–7], as being important for T cell receptor signaling in CTLs. We demonstrate that cytolytic granules are not only reservoirs of cytolytic proteins but are also the acidic Ca2+ stores mobilized by NAADP via TPC channels on the granules themselves, so that TPCs migrate to the immunological synapse upon CTL activation. Moreover, NAADP activates TPCs to drive exocytosis in a way that is not mimicked by global Ca2+ signals induced by IP3 or ionomycin, suggesting that critical, local Ca2+ nanodomains around TPCs stimulate granule exocytosis. Hence, by virtue of the NAADP/TPC pathway, cytolytic granules generate Ca2+ signals that lead to their own exocytosis and to cell killing. This study highlights a selective role for NAADP in stimulating exocytosis crucial for immune cell function and may impact on stimulus-secretion coupling in wider cellular contexts.
Collapse
Affiliation(s)
- Lianne C Davis
- Department of Pharmacology, University of Oxford, Oxford, UK.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Generation of HCMV-specific T-cell Lines From Seropositive Solid-organ-transplant Recipients for Adoptive T-cell Therapy. J Immunother 2009; 32:932-40. [DOI: 10.1097/cji.0b013e3181b88fda] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
7
|
Shanker A, Brooks AD, Jacobsen KM, Wine JW, Wiltrout RH, Yagita H, Sayers TJ. Antigen presented by tumors in vivo determines the nature of CD8+ T-cell cytotoxicity. Cancer Res 2009; 69:6615-23. [PMID: 19654302 DOI: 10.1158/0008-5472.can-09-0685] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The biological relevance of the perforin and Fas ligand (FasL) cytolytic pathways of CD8(+) T lymphocytes (CTL) for cancer immunotherapy is controversial. We investigated the importance of these pathways in a murine renal cell carcinoma expressing influenza viral hemagglutinin as a defined surrogate antigen (Renca-HA). Following Renca-HA injection, all FasL-dysfunctional FasL(gld/gld) mice (n = 54) died from Renca-HA tumors by day 62. By contrast, perforin(-/-) (51%; n = 45) and Fas(lpr/lpr) (55%; n = 51) mice remained tumor-free at day 360. Blocking FasL in vivo inhibited tumor rejection in these mice. Moreover, established Renca-HA tumors were cleared more efficiently by adoptively transferred HA(518-526)-specific T-cell receptor-transgenic CTL using FasL rather than perforin. Strikingly, a range of mouse tumor cells presenting low concentrations of immunogenic peptide were all preferentially lysed by the FasL but not the Pfp-mediated effector pathway of CTL, whereas at higher peptide concentrations, the preference in effector pathway usage by CTL was lost. Interestingly, a number of human renal cancer lines were also susceptible to FasL-mediated cytotoxicity. Therefore, the FasL cytolytic pathway may be particularly important for eradicating Fas-sensitive tumors presenting low levels of MHC class I-associated antigens following adoptive T-cell therapy.
Collapse
Affiliation(s)
- Anil Shanker
- Laboratory of Experimental Immunology, Cancer and Inflammation Program, National Cancer Institute-Frederick, Maryland 21702, USA.
| | | | | | | | | | | | | |
Collapse
|
8
|
Sarcon AK, Desierto MJ, Zhou W, Visconte V, Gibellini F, Chen J, Young NS. Role of perforin-mediated cell apoptosis in murine models of infusion-induced bone marrow failure. Exp Hematol 2009; 37:477-86. [PMID: 19216020 DOI: 10.1016/j.exphem.2008.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2008] [Revised: 11/26/2008] [Accepted: 12/02/2008] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To investigate the role of perforin-mediated cell apoptosis in murine models of immune-mediated bone marrow (BM) failure. MATERIALS AND METHODS We compared C57BL/6J (B6) mice carrying a perforin gene deletion (Prf(-/-)) with wild-type (WT) controls for cellular composition in lymphohematopoietic tissues. Lymph node (LN) cells from Prf(-/-) mice were coincubated with BM cells from B10-H2(b)/LilMcdJ (C.B10) mice in an apoptosis assay in vitro. We then infused Prf(-/-) and WT B6 LN cells into sublethally irradiated C.B10 and CByB6F1 recipients with mismatches at the minor and major histocompatibility loci, respectively, in order to induce BM failure. Cellular composition was analyzed by flow cytometry. RESULTS Prf(-/-) mice showed normal lymphoid cell composition, but Prf(-/-) LN cells had reduced ability to induce C.B10 BM cell apoptosis in vitro. Infusion of 5 to 10 x 10(6) Prf(-/-) LN cells produced obvious BM failure in C.B10 and CByB6F1 recipients; pancytopenia and BM hypocellularity were only slightly less severe than those caused by infusion of 5 x 10(6) WT B6 LN cells. Infused Prf(-/-) LN cells showed less T-cell expansion, normal T-cell activation, and higher proportions of T cells expressing gamma-interferon, tissue necrosis factor-alpha, and Fas ligand CD178, in comparison to infused WT B6 LN cells. Fas expression was equally high in residual BM cells in recipient of both Prf(-/-) and B6 LN cells. CONCLUSION Perforin deficiency alters T-cell expansion but upregulates T-cell Fas ligand expression. Perforin-mediated cell death appears to play a minor role in mouse models of immune-mediated BM failure.
Collapse
Affiliation(s)
- Annahita K Sarcon
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1202, USA
| | | | | | | | | | | | | |
Collapse
|
9
|
He JS, Ostergaard HL. CTLs contain and use intracellular stores of FasL distinct from cytolytic granules. THE JOURNAL OF IMMUNOLOGY 2007; 179:2339-48. [PMID: 17675495 DOI: 10.4049/jimmunol.179.4.2339] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CTL lyse target cells through the release of cytolytic granule contents and cell surface expression of Fas ligand (FasL). Current models suggest that FasL is stored in cytolytic granules and that FasL cell surface expression would be subject to the same controls as degranulation. We demonstrate that murine CTLs undergo two waves of FasL cell surface expression after stimulation. The first wave is from a pre-existing pool of FasL, and the second wave requires new protein synthesis. Signaling for FasL expression appears to be finely tuned as a weak signal preferentially induced surface translocation of the stored FasL, whereas a strong signal preferentially triggered the expression of de novo synthesized FasL. The early FasL is differentially regulated from degranulation, as there were multiple circumstances whereby rapid FasL cell surface expression and FasL-dependent killing occurred in the absence of detectable degranulation. Furthermore, we found through confocal microscopy that stored FasL resides in vesicles distinct from cytolytic granules. Our data clearly show that CTL degranulation and FasL lytic mechanisms are fully independent with respect to stored component localization and regulation.
Collapse
Affiliation(s)
- Jin-Shu He
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Canada
| | | |
Collapse
|
10
|
Yang D, Ud Din N, Browning DD, Abrams SI, Liu K. Targeting lymphotoxin beta receptor with tumor-specific T lymphocytes for tumor regression. Clin Cancer Res 2007; 13:5202-10. [PMID: 17785576 DOI: 10.1158/1078-0432.ccr-07-1161] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE One of the impediments of immunotherapy against cancer is the suppression of tumor-specific CTLs in the tumor microenvironment, partly due to the selective inhibition of the perforin pathway and the emergence of Fas-resistant tumors. Therefore, we sought to identify perforin- and Fas-independent cytotoxic pathways and explored the potential of targeting LTbetaR with tumor-specific CTLs to induce tumor rejection in vivo. EXPERIMENTAL DESIGN Fas-resistant tumors were examined for their susceptibility to perforin-deficient (pfp) CTLs via CTL adoptive transfer in mouse models of experimental lung metastasis. The specificity of LTbetaR, a cell surface death receptor, in causing tumor rejection by CTLs was analyzed by LTbetaR-specific neutralizing monoclonal antibody in vitro. The specificity and efficacy of LTbetaR in the suppression of established tumors was further investigated by silencing LTbetaR in tumor cells in vivo. RESULTS pfp CTLs exhibited significant cytotoxicity against Fas-resistant tumors in vivo. The perforin- and Fas-independent cytotoxicity was directly mediated, at least in part, by the adoptively transferred CTLs. It was observed that LTbetaR was expressed on the tumor cell surface, and LTalpha, LTbeta, and LIGHT, all of which are ligands for LTbetaR, were either constitutively expressed or activated in the tumor-specific CTLs and primary CD8(+) T cells. Blocking LTbetaR with LTbetaR-specific neutralizing monoclonal antibody decreased CTL cytotoxicity in vitro. Silencing LTbetaR using LTbetaR-specific short hairpin RNA reduced the ability of pfp CTLs to induce tumor rejection in vivo. CONCLUSION LTbetaR directly mediates CTL-directed tumor rejection in vivo. Targeting LTbetaR with tumor-specific CTLs is a potential therapeutic approach.
Collapse
Affiliation(s)
- Dafeng Yang
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, Georgia 30912, USA
| | | | | | | | | |
Collapse
|
11
|
de Hooge ASK, Berghuis D, Santos SJ, Mooiman E, Romeo S, Kummer JA, Egeler RM, van Tol MJD, Melief CJM, Hogendoorn PCW, Lankester AC. Expression of Cellular FLICE Inhibitory Protein, Caspase-8, and Protease Inhibitor-9 in Ewing Sarcoma and Implications for Susceptibility to Cytotoxic Pathways. Clin Cancer Res 2007; 13:206-14. [PMID: 17200356 DOI: 10.1158/1078-0432.ccr-06-1457] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Ewing sarcoma is a common pediatric bone tumor with an unfavorable prognosis for metastatic or recurrent disease. Cellular immunotherapy may provide new treatment options and depends on the cytolytic death receptor and perforin/granzyme pathways. Expression of death receptor pathway inhibitor cellular FLICE inhibitory protein (cFLIP), initiator caspase-8, and granzyme B inhibitor protease inhibitor-9 (PI-9) have been reported to determine susceptibility to cell- and chemotherapy-mediated killing in several tumor types. Here, we have studied their in vitro and in vivo expression in Ewing sarcoma and the implications for susceptibility to cytotoxicity. EXPERIMENTAL DESIGN Ewing sarcoma cell lines (n = 8) were tested for cFLIP, PI-9, and caspase-8 expression. Functional significance was tested by anti-Fas antibody (death receptor pathway) or natural killer cell (perforin/granzyme pathway) treatment. Immunohistochemistry was done on 28 sections from 18 patients. In half of the cases, sequential material, including metastases, was available. RESULTS Although all tested Ewing sarcoma cell lines expressed cFLIP, resistance to CD95/Fas-mediated apoptosis was only observed in two cell lines lacking caspase-8 expression. PI-9 was expressed at low levels in four of eight Ewing sarcoma cell lines, but positive cell lines remained susceptible to perforin/granzyme-mediated killing. In primary Ewing sarcoma, including metastases, cFLIP was abundantly expressed in 18 of 18 patients. Caspase-8 was expressed in all patients but showed more intertumoral and intratumoral variation in both intensity and heterogeneity of staining. PI-9, in contrast, was undetectable. CONCLUSIONS The expression patterns of cFLIP, caspase-8, and the absence of PI-9 provide a rationale to preferentially exploit the perforin/granzyme pathway in cytotoxic therapies against Ewing sarcoma.
Collapse
Affiliation(s)
- Alfons S K de Hooge
- Department of Pediatrics, Division of Immunology, Haematology, Oncology, Bone Marrow Transplantation and Autoimmune Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Abdool K, Cretney E, Brooks AD, Kelly JM, Swann J, Shanker A, Bere EW, Yokoyama WM, Ortaldo JR, Smyth MJ, Sayers TJ. NK cells use NKG2D to recognize a mouse renal cancer (Renca), yet require intercellular adhesion molecule-1 expression on the tumor cells for optimal perforin-dependent effector function. THE JOURNAL OF IMMUNOLOGY 2006; 177:2575-83. [PMID: 16888019 DOI: 10.4049/jimmunol.177.4.2575] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The NKG2D receptor on NK cells can recognize a variety of ligands on the tumor cell surface. Using a mouse renal cancer (Renca), we show that NKG2D recognition by NK cells was crucial for their ability to limit tumor metastases in vivo in both liver and lungs using perforin-dependent effector mechanisms. However, for the R331 cell line established from Renca, NKG2D recognition and perforin-dependent lysis played no role in controlling liver metastases. R331 cells were also more resistant to perforin-dependent lysis by NK cells in vitro. We therefore used these phenotypic differences between Renca and R331 to further investigate the crucial receptor:ligand interactions required for triggering lytic effector functions of NK cells. Reconstitution of R331 cells with ICAM-1, but not Rae-1gamma, restored NKG2D-mediated, perforin-dependent lysis. Interestingly, R331 cells were efficiently lysed by NK cells using death ligand-mediated apoptosis. This death ligand-mediated killing did not depend on NKG2D recognition of its ligands on tumor cells. This result suggests that the intracellular signaling in NK cells required for perforin and death ligand-mediated lysis of tumor target cell are quite distinct, and activation of both of these antitumor lytic effector functions of NK cells could improve therapeutic benefits for certain tumors.
Collapse
MESH Headings
- Animals
- Carcinoma, Renal Cell/immunology
- Carcinoma, Renal Cell/metabolism
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/secondary
- Cell Line, Tumor
- Cells, Cultured
- Cytotoxicity, Immunologic/genetics
- Intercellular Adhesion Molecule-1/biosynthesis
- Intercellular Adhesion Molecule-1/genetics
- Kidney Neoplasms/immunology
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/pathology
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Liver Neoplasms, Experimental/immunology
- Liver Neoplasms, Experimental/metabolism
- Liver Neoplasms, Experimental/pathology
- Liver Neoplasms, Experimental/secondary
- Membrane Proteins/deficiency
- Membrane Proteins/genetics
- Membrane Proteins/physiology
- Mice
- Mice, Inbred BALB C
- NK Cell Lectin-Like Receptor Subfamily K
- Pore Forming Cytotoxic Proteins
- Receptors, Immunologic/physiology
- Receptors, Natural Killer Cell
Collapse
Affiliation(s)
- Karen Abdool
- Laboratory of Experimental Immunology, National Cancer Institute-Frederick, Building 560, Frederick, MD 21702, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Liu K, Caldwell SA, Greeneltch KM, Yang D, Abrams SI. CTL adoptive immunotherapy concurrently mediates tumor regression and tumor escape. THE JOURNAL OF IMMUNOLOGY 2006; 176:3374-82. [PMID: 16517705 DOI: 10.4049/jimmunol.176.6.3374] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Tumor escape and recurrence are major impediments for successful immunotherapy. It is well-documented that the emergence of Ag-loss variants, as well as regulatory mechanisms suppressing T cell function, have been linked to inadequate antitumor activity. However, little is known regarding the role of Fas-mediated cytotoxicity by tumor-specific CD8(+) CTL in causing immune evasion of Fas resistant variants during adoptive immunotherapy. In this study, we made use of an adoptive transfer model of experimental lung metastasis using tumor-specific CTL as a relevant immune-based selective pressure, and wherein the Fas ligand pathway was involved in the antitumor response. Surviving tumor cells were recovered and examined for alterations in antigenic, functional, and biologic properties. We showed that diminished susceptibility to Fas-mediated cytotoxicity in vivo was an important determinant of tumor escape following CTL-based immunotherapy. Tumor escape variants (TEV) recovered from the lungs of CTL-treated mice exhibited more aggressive behavior in vivo. However, these TEV retained relevant MHC class I and tumor Ag expression and sensitivity to CTL via the perforin pathway but reduced susceptibility to Fas-mediated lysis. Moreover, TEV were significantly less responsive to eradication by CTL adoptive immunotherapy paradigms as a consequence of increased Fas resistance. Overall, we identified that Fas(low)-TEV emerged as a direct consequence of CTL-tumor interactions in vivo, and that such an altered neoplastic Fas phenotype compromised immunotherapy efficacy. Together, these findings may have important implications for both tumor progression and the design of immunotherapeutic interventions to confront these selective pressures or escape mechanisms.
Collapse
Affiliation(s)
- Kebin Liu
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bldg. 10, 10 Center Drive, Bethesda, MD 20892, USA.
| | | | | | | | | |
Collapse
|
14
|
Atherly LO, Brehm MA, Welsh RM, Berg LJ. Tec kinases Itk and Rlk are required for CD8+ T cell responses to virus infection independent of their role in CD4+ T cell help. THE JOURNAL OF IMMUNOLOGY 2006; 176:1571-81. [PMID: 16424186 DOI: 10.4049/jimmunol.176.3.1571] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Itk and Rlk are members of the Tec kinase family of nonreceptor protein tyrosine kinases that are expressed in T cells, NK cells, and mast cells. These proteins are involved in the regulation of signaling processes downstream of the TCR in CD4(+) T cells, particularly in the phosphorylation of phospholipase C-gamma1 after TCR activation; furthermore, both Itk and Rlk are important in CD4(+) T cell development, differentiation, function, and homeostasis. However, few studies have addressed the roles of these kinases in CD8(+) T cell signaling and function. Using Itk(-/-) and Itk(-/-)Rlk(-/-) mice, we examined the roles of these Tec family kinases in CD8(+) T cells, both in vitro and in vivo. These studies demonstrate that the loss of Itk and Rlk impairs TCR-dependent signaling, causing defects in phospholipase C-gamma1, p38, and ERK activation as well as defects in calcium flux and cytokine production in vitro and expansion and effector cytokine production by CD8(+) T cells in response to viral infection. These defects cannot be rescued by providing virus-specific CD4(+) T cell help, thereby substantiating the important role of Tec kinases in CD8(+) T cell signaling.
Collapse
Affiliation(s)
- Luana O Atherly
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | | | | | |
Collapse
|
15
|
Gallo EM, Canté-Barrett K, Crabtree GR. Lymphocyte calcium signaling from membrane to nucleus. Nat Immunol 2005; 7:25-32. [PMID: 16357855 DOI: 10.1038/ni1295] [Citation(s) in RCA: 151] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2005] [Accepted: 11/14/2005] [Indexed: 12/14/2022]
Abstract
Ca(2+) signals control a variety of lymphocyte responses, ranging from short-term cytoskeletal modifications to long-term changes in gene expression. The identification of molecules and channels that modulate Ca(2+) entry into T and B lymphocytes has both provided details of the molecular events leading to immune responses and raised controversy. Here we review studies of the pathways that allow Ca(2+) entry, the function of Ca(2+) in the regulation of cell polarity and motility and the principles by which Ca(2+)-dependent transcription regulates lymphocyte function.
Collapse
Affiliation(s)
- Elena M Gallo
- Program in Immunology, Stanford University, Stanford, California 94305, USA
| | | | | |
Collapse
|
16
|
Stuart PM, Yin X, Plambeck S, Pan F, Ferguson TA. The role of Fas ligand as an effector molecule in corneal graft rejection. Eur J Immunol 2005; 35:2591-7. [PMID: 16114107 DOI: 10.1002/eji.200425934] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Previous studies have shown that the expression of Fas ligand (FasL; CD95L) by donor corneas is critical to their survival when placed on allogeneic recipients. Since there have been reports that the cornea expresses Fas, we tested the idea that FasL on lymphoid cells could be an effector molecule during rejection episodes. When FasL defective BALB/c-gld mice were engrafted with allogeneic corneas, significantly more of these corneas were accepted than by normal BALB/c mice. However, this was not due to impaired FasL-mediated effector function in these mice as the allogeneic corneas did not express detectable Fas by Western blot or RT-PCR analysis. Furthermore, donor corneas without Fas were given no survival advantage, but were rejected similar to wild-type donor allogeneic corneas. Examination of the T cell compartment in gld mice revealed that these cells express higher levels of Fas and are more susceptible to Fas-mediated death than wild-type cells. These results indicate that FasL is not an effector molecule in corneal graft rejection and that gld mice show reduced graft rejection due to greater susceptibility of their T cells to Fas-mediated apoptosis.
Collapse
Affiliation(s)
- Patrick M Stuart
- Department of Ophthalmology and Visual Sciences, Washington University Medical School, St. Louis, MO 63110, USA.
| | | | | | | | | |
Collapse
|
17
|
Lin MY, Zal T, Ch'en IL, Gascoigne NRJ, Hedrick SM. A pivotal role for the multifunctional calcium/calmodulin-dependent protein kinase II in T cells: from activation to unresponsiveness. THE JOURNAL OF IMMUNOLOGY 2005; 174:5583-92. [PMID: 15843557 DOI: 10.4049/jimmunol.174.9.5583] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Stimulation of the TCR leads to an oscillatory release of free calcium that activates members of the calcium/calmodulin-dependent protein kinase II (CaMKII) family. The CaMKII molecules have profound and lasting effects on cellular signaling in several cell types, yet the role of CaMKII in T cells is still poorly characterized. In this report we describe a splice variant of CaMKIIbeta, CaMKIIbeta'e, in mouse T cells. We have determined its function, along with that of CaMKIIgamma, by introducing the active and kinase-dead mutants into activated P14 TCR transgenic T cells using retroviral transduction. Active CaMKII enhanced the proliferation and cytotoxic activity of T cells while reducing their IL-2 production. Furthermore, it induced a profound state of unresponsiveness that could be overcome only by prolonged culture in IL-2. These results indicate that members of the CaMKII family play an important role in regulation of CD8 T cell proliferation, cytotoxic effector function, and the response to restimulation.
Collapse
Affiliation(s)
- Meei Yun Lin
- Division of Biological Sciences, The Cancer Center, and Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, CA 92093, USA
| | | | | | | | | |
Collapse
|
18
|
Jones SC, Friedman TM, Murphy GF, Korngold R. Specific donor Vβ-associated CD4+ T-cell responses correlate with severe acute graft-versus-host disease directed to multiple minor histocompatibility antigens. Biol Blood Marrow Transplant 2004; 10:91-105. [PMID: 14750075 DOI: 10.1016/j.bbmt.2003.10.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CXB-2/By (CXB-2) recombinant inbred mice express a subset of the minor histocompatibility antigen (miHA) repertoire expressed by C.B10-H2(b)/LiMcdJ (BALB.B) mice. On lethal irradiation and the transplantation of H2(b)-matched C57BL/6 (B6) T cell-depleted bone marrow cells, along with naive unfractionated T cells, both strains succumb to acute graft-versus-host disease (GVHD). Although alloreactive B6 CD4(+) T cells are a necessary source of T-cell help for the B6 CD8(+) component of the GVHD response in both recipient strains, they are capable of mediating severe GVHD by themselves only in BALB.B mice. Previous CD4(+) T-cell receptor repertoire analysis demonstrated overlapping oligoclonal Vbeta use between the CD4(+) B6 anti-BALB.B and B6 anti-CXB-2 responses, with indications of additional BALB.B unique T-cell responses (Vbeta2 and Vbeta11). We report here that the more severe B6 anti-BALB.B response is not due to a quantitative difference in the responding cells, because the frequency of alloreactive donor CD4(+) T cells over time was equivalent in the spleens of BALB.B versus CXB-2 recipients. The responses were also similar in the number of infiltrating B6 CD4(+) T cells in the lingual epithelium of the 2 recipients. In contrast, a significantly greater degree of infiltration and injury of BALB.B intestinal epithelium correlated with the increased level of clinical GVHD severity. Of most significance, despite the involvement of at least 11 Vbeta-associated CD4(+) T-cell families in the overall B6 anti-BALB.B response, the development of severe GVHD correlated with the presence of Vbeta2- and Vbeta11-positive donor T cells. Transplantation of donor CD4(+) T cells from Vbeta-associated families that were shared between the B6 anti-BALB.B and anti-CXB-2 responses resulted in minimal GVHD potential. These data suggest that severe GVHD across miHA barriers depends on the involvement of a restricted number of potent T-cell specificities and implies that there are only a limited number of corresponding responsible miHAs.
Collapse
Affiliation(s)
- Stephen C Jones
- The Kimmel Cancer Center, Jefferson Medical College, Philadelphia, PA 19107, USA
| | | | | | | |
Collapse
|
19
|
Kafrouni MI, Brown GR, Thiele DL. The role of TNF-TNFR2 interactions in generation of CTL responses and clearance of hepatic adenovirus infection. J Leukoc Biol 2003; 74:564-71. [PMID: 12960267 DOI: 10.1189/jlb.0103035] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Deficiency or inhibition of tumor necrosis factor (TNF) significantly prolongs hepatic expression of recombinant adenoviral vectors. To explore mechanisms responsible for this observation, the present studies examined the effects of TNF versus TNF receptor 1 (TNFR1) or TNFR2 deficiency on the course of antiviral-immune responses to a replication-deficient, beta-galactosidase-encoding recombinant adenovirus (AdCMV-lacZ). Clearance of AdCMV-lacZ was significantly delayed in TNF-deficient mice. Less pronounced but significant delays in AdCMV-lacZ clearance were observed in TNFR2-deficient but not TNFR1-deficient mice. Numbers of interferon-gamma expressing intrahepatic lymphocytes (IHL) were similar in AdCMV-lacZ-infected, TNF-deficient, TNFR1-deficient, TNFR2-deficient, and control mice. However, IHL isolated from AdCMV-lacZ-infected, TNF-deficient or AdCMV-lacZ-infected, TNFR2-deficient mice exhibited decreased levels of FasL expression and adenovirus-specific cytolytic T lymphocyte (CTL) activity. Similar defects in allo-specific killing of Fas-sensitive hepatocyte targets by TNF-deficient or TNFR2-deficient but not TNFR1-deficient CTL were also noted. No defects in generation of allo-specific cytotoxicity directed against perforin-sensitive target cells were noted in TNF-, TNFR1-, or TNFR2-deficient lymphocytes. These findings indicate that TNF/TNFR2 interactions facilitate generation of FasL-dependent CTL effector pathways that play an important role in in vivo antiviral-immune responses in the liver.
Collapse
MESH Headings
- Adenoviridae/immunology
- Animals
- Antigens, CD/physiology
- Cytotoxicity, Immunologic
- Fas Ligand Protein
- Interferon-gamma/analysis
- Liver/immunology
- Liver/virology
- Membrane Glycoproteins/analysis
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Receptors, Tumor Necrosis Factor/physiology
- Receptors, Tumor Necrosis Factor, Type I
- Receptors, Tumor Necrosis Factor, Type II
- T-Lymphocytes, Cytotoxic/immunology
- Tumor Necrosis Factor-alpha/physiology
Collapse
Affiliation(s)
- Michel I Kafrouni
- Department of Internal Medicine, Divison of Digestive and Liver Diseases, University of Texas Southwestern Medical Center at Dallas, 75390-9151, USA
| | | | | |
Collapse
|
20
|
Caldwell SA, Ryan MH, McDuffie E, Abrams SI. The Fas/Fas ligand pathway is important for optimal tumor regression in a mouse model of CTL adoptive immunotherapy of experimental CMS4 lung metastases. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 171:2402-12. [PMID: 12928387 DOI: 10.4049/jimmunol.171.5.2402] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The mechanisms of CTL-mediated tumor regression in vivo remain to be fully understood. If CTL do mediate tumor regression in vivo by direct cytotoxicity, this may occur via two major effector mechanisms involving the secretion of perforin/granzymes and/or engagement of Fas by Fas ligand (FasL) expressed by the activated CTL. Although the perforin pathway has been considered the dominant player, it is unclear whether Fas-mediated cytotoxicity is additionally required for optimal tumor rejection. Previously, we produced H-2L(d)-restricted CTL reactive against the CMS4 sarcoma, which expresses a naturally occurring rejection Ag recognized by these CTL and harbors a cytokine (IFN-gamma plus TNF)-inducible, Fas-responsive phenotype. The adoptive transfer of these CTL to syngeneic BALB/c mice with minimal (day 3 established) or extensive (day 10 established) experimental pulmonary metastases resulted in strong antitumor responses. Here we investigated whether a FasL-dependent CTL effector mechanism was important for optimal tumor regression in this adoptive immunotherapy model. The approach taken was to compare the therapeutic efficacy of wild-type to FasL-deficient (gld) CTL clones by adoptive transfer. In comparison with wild-type CTL, gld-CTL efficiently mediated tumor cytolysis and produced comparable amounts of IFN-gamma, after tumor-specific stimulation, as in vitro assessments of Ag recognition. Moreover, gld-CTL mediated comparably potent antitumor effects in a minimal disease setting, but were significantly less effective under conditions of an extensive tumor burden. Overall, under conditions of extensive lung metastases, these data revealed for the first time an important role for a FasL-dependent CTL effector mechanism in optimal tumor regression.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Clone Cells
- Cytotoxicity, Immunologic/genetics
- Disease Models, Animal
- Fas Ligand Protein
- Female
- Immunotherapy, Adoptive/methods
- Injections, Intravenous
- Ligands
- Lung Neoplasms/genetics
- Lung Neoplasms/immunology
- Lung Neoplasms/secondary
- Lung Neoplasms/therapy
- Lymphocyte Activation/genetics
- Membrane Glycoproteins/deficiency
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/physiology
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Neoplasm Transplantation
- Perforin
- Pore Forming Cytotoxic Proteins
- Sarcoma, Experimental/genetics
- Sarcoma, Experimental/immunology
- Sarcoma, Experimental/secondary
- Sarcoma, Experimental/therapy
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Cytotoxic/transplantation
- fas Receptor/biosynthesis
- fas Receptor/physiology
Collapse
Affiliation(s)
- Sheila A Caldwell
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
21
|
Affiliation(s)
- Pere Santamaria
- Department of Microbiology and Infectious Diseases and Julia McFarlane Diabetes Research Centre, Faculty of Medicine, The University of Calgary, 3330 Hospital Drive N.W., Calgary T2N 4N1, Canada.
| |
Collapse
|
22
|
Lim DG, Slavik JM, Bourcier K, Smith KJ, Hafler DA. Allelic variation of MHC structure alters peptide ligands to induce atypical partial agonistic CD8+ T cell function. J Exp Med 2003; 198:99-109. [PMID: 12847139 PMCID: PMC2196091 DOI: 10.1084/jem.20021796] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
T cell receptors recognize small changes in peptide ligands leading to different T cell responses. Here, we analyzed a panel of HLA-A2-Tax11-19 reactive T cell clones to examine how small allelic variations of MHC molecules could alter the functional outcome of antigen recognition. Similar to the effects induced by antigenic altered peptide ligands, weak or partial agonistic T cell functions were identified in individual T cell clones with the recognition of MHC-altered peptide ligands (MAPLs). Interestingly, one subtype of HLA-A2 molecules induced an unusual type of partial agonistic function; proliferation without cytotoxicity. Modeling of crystallographic data indicated that polymorphic amino acids in the HLA-A2 peptide binding groove, especially the D-pocket, were responsible for this partial agonism. Reciprocal mutations of the Tax peptide side chain engaging the D-pocket indeed restored the agonist functions of the MHC-peptide complex. Whereas early intracellular signaling events were not efficiently induced by these MAPLs, phosphorylated c-Jun slowly accumulated with sustained long-term expression. These data indicate that MAPLs can induce atypical partial agonistic T cell function through structural and biochemical mechanisms similar to altered peptide ligands.
Collapse
Affiliation(s)
- Dong-Gyun Lim
- Laboratory of Molecular Immunology, Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Institutes of Medicine, Boston, MA 02115-5817, USA
| | | | | | | | | |
Collapse
|
23
|
Abstract
The aim of this study was to examine in detail the low functional capacity of human intraepithelial lymphocytes (IELs) in response to phytohaemagglutinin (PHA) and CD3 ligation. Human IELs were extracted from jejunal mucosa obtained from patients undergoing gastric bypass operations for morbid obesity and compared to peripheral blood (PB) lymphocytes composed predominantly of CD8+ T cells. Calcium influx ([Ca2+]i) was analysed using Fura-2-loaded cells; IL-2 receptor expression was measured by immunofluorescence and flow cytometry; IL-2 binding was determined using radiolabelled IL-2; IL-2 production was quantified by ELISA; and apoptosis was detected with Apo 2.7 staining. Compared to naive PB CD8+ T lymphocytes, calcium influx by IELs was only transient with CD3 ligation and low in amplitude with PHA. IL-2 receptor expression was reduced after CD3 ligation, yet normal in numbers and affinity after PHA stimulation. Both cell types secreted similar amounts of IL-2. CD3 expression on IELs, but not PB CD8+ T cells, declined upon activation, due partly to incomplete reexpression after modulation. Little apoptosis was found. The partial activation of IELs in response to PHA and CD3 ligation, as manifested by diminished [Ca2+]i, resulted in a decline in CD3 expression.
Collapse
Affiliation(s)
- E C Ebert
- Department of Medicine, UMDNJ-Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| |
Collapse
|
24
|
Guillaume P, Legler DF, Boucheron N, Doucey MA, Cerottini JC, Luescher IF. Soluble major histocompatibility complex-peptide octamers with impaired CD8 binding selectively induce Fas-dependent apoptosis. J Biol Chem 2003; 278:4500-9. [PMID: 12407102 DOI: 10.1074/jbc.m208863200] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Fluorescence-labeled soluble major histocompatibility complex class I-peptide "tetramers" constitute a powerful tool to detect and isolate antigen-specific CD8(+) T cells by flow cytometry. Conventional "tetramers" are prepared by refolding of heavy and light chains with a specific peptide, enzymatic biotinylation at an added C-terminal biotinylation sequence, and "tetramerization" by reaction with phycoerythrin- or allophycocyanin-labeled avidin derivatives. We show here that such preparations are heterogeneous and describe a new procedure that allows the preparation of homogeneous tetra- or octameric major histocompatibility complex-peptide complexes. These compounds were tested on T1 cytotoxic T lymphocytes (CTLs), which recognize the Plasmodium berghei circumsporzoite peptide 252-260 (SYIPSAEKI) containing photoreactive 4-azidobenzoic acid on Lys(259) in the context of H-2K(d). We report that mutation of the CD8 binding site of K(d) greatly impairs the binding of tetrameric but not octameric or multimeric K(d)-PbCS(ABA) complexes to CTLs. This mutation abolishes the ability of the octamer to elicit significant phosphorylation of CD3, intracellular calcium mobilization, and CTL degranulation. Remarkably, however, this octamer efficiently activates CTLs for Fas (CD95)-dependent apoptosis.
Collapse
Affiliation(s)
- Philippe Guillaume
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, 1066 Epalinges, Switzerland
| | | | | | | | | | | |
Collapse
|
25
|
Le Roy E, Baron M, Faigle W, Clément D, Lewinsohn DM, Streblow DN, Nelson JA, Amigorena S, Davignon JL. Infection of APC by human cytomegalovirus controlled through recognition of endogenous nuclear immediate early protein 1 by specific CD4(+) T lymphocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:1293-301. [PMID: 12133951 DOI: 10.4049/jimmunol.169.3.1293] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Infections by human CMV are controlled by cellular immune responses. Professional APC such as monocytes and macrophages can be infected in vivo and are considered as a reservoir of virus. However, CMV-specific CD4(+) responses against infected APC have not been reported. To develop a model of CD4-infected APC interaction, we have transfected the U373MG astrocytoma cell line with the class II transactivator (CIITA). Confocal microscopy experiments showed that U373MG-CIITA cells expressed markers characteristic of APC. Functional assays demonstrated that infected U373MG-CIITA APC processed and presented both exogenous and endogenously neosynthesized nuclear immediate early (IE) protein 1 through the MHC class II pathway. More importantly, endogenous presentation of IE1 by infected APC lead to efficient control of CMV infection as revealed by decreased viral titer. Thus, these results describe the endogenous presentation of a nuclear viral protein by the MHC class II pathway and suggest that IE1-specific CD4(+) T cells may play an important role in CMV infection by directly acting against infected APC.
Collapse
Affiliation(s)
- Emmanuelle Le Roy
- Institut National de la Santé et de la Recherche Médicale, Toulouse, France
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Seki N, Brooks AD, Carter CRD, Back TC, Parsoneault EM, Smyth MJ, Wiltrout RH, Sayers TJ. Tumor-specific CTL kill murine renal cancer cells using both perforin and Fas ligand-mediated lysis in vitro, but cause tumor regression in vivo in the absence of perforin. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:3484-92. [PMID: 11907109 DOI: 10.4049/jimmunol.168.7.3484] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Kidney cancer is a devastating disease; however, biological therapies have achieved some limited success. The murine renal cancer Renca has been used as a model for developing new preclinical approaches to the treatment of renal cell carcinoma. Successful cytokine-based approaches require CD8(+) T cells, but the exact mechanisms by which T cells mediate therapeutic benefit have not been completely identified. After successful biological therapy of Renca in BALB/c mice, we generated CTLs in vitro using mixed lymphocyte tumor cultures. These CTL mediated tumor-specific H-2K(d)-restricted lysis and production of IFN-gamma, TNF-alpha, and Fas ligand (FasL) in response to Renca. CTL used both granule- and FasL-mediated mechanisms to lyse Renca, although granule-mediated killing was the predominant lytic mechanism in vitro. The cytokines IFN-gamma and TNF-alpha increased the sensitivity of Renca cells to CTL lysis by both granule- and FasL-mediated death pathways. Adoptive transfer of these anti-Renca CTL into tumor-bearing mice cured most mice of established experimental pulmonary metastases, and successfully treated mice were immune to tumor rechallenge. Interestingly, we were able to establish Renca-specific CTL from mice gene targeted for perforin (pfp(-/-)) mice. Although these pfp(-/-) CTL showed reduced cytotoxic activity against Renca, their IFN-gamma production in the presence of Renca targets was equivalent to that of wild-type CTL, and adoptive transfer of pfp(-/-) CTL was as efficient as wild-type CTL in causing regression of established Renca pulmonary metastases. Therefore, although granule-mediated killing is of paramount importance for CTL-mediated lysis in vitro, some major in vivo effector mechanisms clearly are independent of perforin.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/toxicity
- Apoptosis/genetics
- Apoptosis/immunology
- Apoptosis Regulatory Proteins
- Carcinoma, Renal Cell/immunology
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/therapy
- Cell Line, Transformed
- Cytotoxicity, Immunologic/genetics
- Epitopes, T-Lymphocyte/administration & dosage
- Epitopes, T-Lymphocyte/immunology
- Fas Ligand Protein
- Immunotherapy, Adoptive/methods
- Intercellular Adhesion Molecule-1/metabolism
- Intercellular Adhesion Molecule-1/physiology
- Kidney Neoplasms/immunology
- Kidney Neoplasms/pathology
- Kidney Neoplasms/therapy
- Ligands
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Lung Neoplasms/secondary
- Lung Neoplasms/therapy
- Lymphocyte Activation/genetics
- Lymphocyte Function-Associated Antigen-1/metabolism
- Lymphocyte Function-Associated Antigen-1/physiology
- Melanoma, Experimental/immunology
- Membrane Glycoproteins/biosynthesis
- Membrane Glycoproteins/deficiency
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Membrane Glycoproteins/toxicity
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Knockout
- Neoplasm Transplantation
- Perforin
- Pore Forming Cytotoxic Proteins
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/transplantation
- TNF-Related Apoptosis-Inducing Ligand
- Tumor Cells, Cultured
- Tumor Necrosis Factor-alpha/toxicity
- fas Receptor/metabolism
Collapse
Affiliation(s)
- Naoko Seki
- Laboratory of Experimental Immunology, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Tau GZ, Cowan SN, Weisburg J, Braunstein NS, Rothman PB. Regulation of IFN-gamma signaling is essential for the cytotoxic activity of CD8(+) T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:5574-82. [PMID: 11698428 PMCID: PMC4416493 DOI: 10.4049/jimmunol.167.10.5574] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Previous studies have demonstrated that, as naive murine CD4(+) cells differentiate into Th1 cells, they lose expression of the second chain of IFN-gammaR (IFN-gammaR2). Hence, the IFN-gamma-producing subset of Th cells is unresponsive to IFN-gamma. Analysis of IFN-gamma-producing CD8(+) T cells demonstrates that, like Th1 cells, these cells do not express IFN-gammaR2. To define the importance of IFN-gamma signaling for the development of functional CD8(+) T cells, mice either lacking IFN-gammaR2 or overexpressing this protein were examined. While CD8(+) T cell development and function appear normal in IFN-gammaR2(-/-) mice, CD8(+) T cell function in IFN-gammaR2 transgenic is altered. IFN-gammaR2 transgenic CD8(+) T cells are unable to lyse target cells in vitro. However, these cells produce Fas ligand, perforin, and granzyme B, the effector molecules required for killing. Interestingly, TG CD8(+) T cells proliferate normally and produce cytokines, such as IFN-gamma in response to antigenic stimulation. Therefore, although IFN-gamma signaling is not required for the generation of normal cytotoxic T cells, constitutive IFN-gamma signaling can selectively impair the cytotoxic function of CD8(+) T cells.
Collapse
MESH Headings
- Animals
- Cell Line
- Cells, Cultured
- Clone Cells
- Cytokines/biosynthesis
- Cytotoxicity Tests, Immunologic
- Cytotoxicity, Immunologic
- Immunologic Memory
- Interferon-gamma/pharmacology
- Lymphocyte Activation
- Mice
- Mice, Knockout
- Mice, Transgenic
- RNA, Messenger/biosynthesis
- Receptors, Interferon/genetics
- Receptors, Interferon/physiology
- Signal Transduction
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- Transcriptional Activation
- Tumor Cells, Cultured
- Interferon gamma Receptor
Collapse
Affiliation(s)
- Gregory Z. Tau
- Integrated Program in Cell, Molecular, and Biophysical Studies, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Simone N. Cowan
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Jeffrey Weisburg
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Ned S. Braunstein
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| | - Paul B. Rothman
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY 10032
- Department of Microbiology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| |
Collapse
|
28
|
Santori FR, Arsov I, Vukmanović S. Modulation of CD8+ T cell response to antigen by the levels of self MHC class I. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:5416-21. [PMID: 11313378 DOI: 10.4049/jimmunol.166.9.5416] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The response of H-Y-specific TCR-transgenic CD8(+) T cells to Ag is characterized by poor proliferation, cytolytic activity, and IFN-gamma secretion. IFN-gamma secretion, but not cytotoxic function, can be rescued by the B7.1 molecule, suggesting that costimulation can selectively enhance some, but not all, effector CD8(+) T cell responses. Although the H-Y epitope binds H-2D(b) relatively less well than some other epitopes, it can induce potent CTL responses in nontransgenic mice, suggesting that the observed poor responsiveness of transgenic CD8(+) T cells cannot be ascribed to the epitope itself. Previously reported reactivity of this TCR to H-2A(b) is also not the cause of the poor responsiveness of the H-Y-specific CD8(+) T cells, as H-Y-specific CD8(+) T cells obtained from genetic backgrounds lacking H-2A(b) also responded poorly. Rather, reducing the levels of H-2(b) class I molecules by breeding the mice to (C57BL/6 x B10.D2)F(1) or TAP1(+/-) backgrounds partially restored cytotoxic activity and enhanced proliferative responses. These findings demonstrate that the self MHC class I gene dosage may regulate the extent of CD8(+) T cell responsiveness to Ag.
Collapse
Affiliation(s)
- F R Santori
- Michael Heidelberger Division of Immunology, Department of Pathology, and Kaplan Comprehensive Cancer Center, New York University School of Medicine, New York, NY 10016, USA
| | | | | |
Collapse
|
29
|
Kreuwel HT, Sherman LA. The role of Fas-FasL in CD8+ T-cell-mediated insulin-dependent diabetes mellitus (IDDM). J Clin Immunol 2001; 21:15-8. [PMID: 11321233 DOI: 10.1023/a:1006780629564] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In the past few years a number of studies have evaluated the contributions of different cytolytic pathways in the autoimmune destruction of pancreatic beta cells, which results in insulin-dependent (type I) diabetes mellitus. Conflicting results continue to emerge regarding the role of Fas-mediated apoptosis in beta-cell destruction. This is likely to reflect differences inherent to the model systems under investigation, as well as the pleiotropic nature of the genes that are involved in cytotoxicity. Despite these complications, it may be possible to reconcile some of these apparently conflicting results by considering that T-cell-mediated cytotoxicity can occur simultaneously by several mechanisms and that variables such as the cytokine milieu and the strength of the signal to the T cell received through the T-cell receptor complex may alter the relative contribution of each cytolytic pathway to beta-cell destruction.
Collapse
Affiliation(s)
- H T Kreuwel
- Department of Immunology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | |
Collapse
|
30
|
Dougall DS, Lamousé-Smith ES, McCarthy SA. Generation and regeneration of a novel anti-CD8-resistant cytolytic T lymphocyte population. Cell Immunol 2000; 205:1-12. [PMID: 11078602 DOI: 10.1006/cimm.2000.1709] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We recently reported that the in vivo development of a novel CD8(+), but anti-CD8 mAb-resistant, CTL population is complex and distinct from that of conventional anti-CD8 mAb-sensitive CD8(+) CTL. In this study, we explored the role of the thymus in the generation of anti-CD8-resistant pCTL and in their maintenance once they are generated. We also investigated the capacities of the adult periphery and thymus to support the regeneration of anti-CD8-resistant pCTL after peripheral lymphocyte and/or thymocyte depletion. These studies indicate that the thymus is necessary for the generation but not the maintenance of peripheral anti-CD8-resistant pCTL. These studies also indicate that the adult thymus can produce these pCTL and the adult periphery can support their regeneration, if a new wave of thymic maturation is experimentally induced. These results may have implications for immune reconstitution after treatment for cancer or HIV infection.
Collapse
Affiliation(s)
- D S Dougall
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 15213, USA
| | | | | |
Collapse
|
31
|
Differential use of FasL- and perforin-mediated cytolytic mechanisms by T-cell subsets involved in graft-versus-myeloid leukemia responses. Blood 2000. [DOI: 10.1182/blood.v96.3.1047.015k36_1047_1055] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
In graft-versus-leukemia (GVL) responses, the cellular subsets and effector mechanisms responsible for cytotoxicity against leukemic cells in vivo remain poorly characterized. A murine model of syngeneic GVL that features CD4+ and CD8+T-cell responses against the MMB3.19 myeloid leukemia cell line has been previously described. MMB3.19 expresses high levels of functional Fas and tumor necrosis factor (TNF) receptors that do not transduce proapoptotic signals. Through the use of perforin- and Fas ligand (FasL)-deficient mice, it was demonstrated that CD4+ T cells mediate anti-MMB3.19 effects in vivo primarily through the use of FasL and secondarily through perforin mechanisms. Conversely, CD8+ T cells induce GVL effects primarily through the use of perforin and minimally through FasL mechanisms. Although the in vivo observations of CD8+ T cells were reflective of their in vitro cytotoxic T lymphocyte (CTL) activity, for CD4+ T cells, in vitro responses were dominated by the perforin pathway. In addition, the diminished capacity of T cells from perforin- and FasL-deficient mice to lyse MMB3.19 target cells appeared directly related to their deficient cytotoxic functions rather than to defects in activation because these cells were fully capable of mounting proliferative responses to the tumor cells. These findings demonstrate that GVL responses of T-cell subsets can involve preferential use of different cytotoxic mechanisms. In particular, these findings identify a role for both FasL-employing CD4+CTLs and the more novel perforin-utilizing CD4+ T-cell subset in responses against a myeloid leukemia.
Collapse
|
32
|
Differential use of FasL- and perforin-mediated cytolytic mechanisms by T-cell subsets involved in graft-versus-myeloid leukemia responses. Blood 2000. [DOI: 10.1182/blood.v96.3.1047] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
In graft-versus-leukemia (GVL) responses, the cellular subsets and effector mechanisms responsible for cytotoxicity against leukemic cells in vivo remain poorly characterized. A murine model of syngeneic GVL that features CD4+ and CD8+T-cell responses against the MMB3.19 myeloid leukemia cell line has been previously described. MMB3.19 expresses high levels of functional Fas and tumor necrosis factor (TNF) receptors that do not transduce proapoptotic signals. Through the use of perforin- and Fas ligand (FasL)-deficient mice, it was demonstrated that CD4+ T cells mediate anti-MMB3.19 effects in vivo primarily through the use of FasL and secondarily through perforin mechanisms. Conversely, CD8+ T cells induce GVL effects primarily through the use of perforin and minimally through FasL mechanisms. Although the in vivo observations of CD8+ T cells were reflective of their in vitro cytotoxic T lymphocyte (CTL) activity, for CD4+ T cells, in vitro responses were dominated by the perforin pathway. In addition, the diminished capacity of T cells from perforin- and FasL-deficient mice to lyse MMB3.19 target cells appeared directly related to their deficient cytotoxic functions rather than to defects in activation because these cells were fully capable of mounting proliferative responses to the tumor cells. These findings demonstrate that GVL responses of T-cell subsets can involve preferential use of different cytotoxic mechanisms. In particular, these findings identify a role for both FasL-employing CD4+CTLs and the more novel perforin-utilizing CD4+ T-cell subset in responses against a myeloid leukemia.
Collapse
|
33
|
Sayers TJ, Brooks AD, Seki N, Smyth MJ, Yagita H, Blazar BR, Malyguine AM. T cell lysis of murine renal cancer: multiple signaling pathways for cell death via Fas. J Leukoc Biol 2000. [DOI: 10.1189/jlb.68.1.81] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
| | - Alan D. Brooks
- Intramural Research Support Program, Frederick, Maryland
| | - Naoko Seki
- Laboratory of Experimental Immunology, DBS, NCI‐FCRDC, Frederick, Maryland
| | - Mark J. Smyth
- Cellular Cytotoxicity Laboratory, The Austin Research Institute, Heidelberg, Victoria, Australia
| | - Hideo Yagita
- Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Bruce R. Blazar
- Department of Pediatrics, Division of Bone Marrow Transplantation, University of Minnesota, Minneapolis
| | | |
Collapse
|
34
|
Soubeyran P, Mallo GV, Moucadel V, Dagorn JC, Iovanna JL. Overexpression of Cdx1 and Cdx2 homeogenes enhances expression of the HLA-I in HT-29 cells. MOLECULAR CELL BIOLOGY RESEARCH COMMUNICATIONS : MCBRC 2000; 3:271-6. [PMID: 10964750 DOI: 10.1006/mcbr.2000.0226] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have previously reported that down-regulation of Cdx1 and Cdx2 mRNA expression is associated with colon carcinogenesis, and that coordinated reexpression of these genes in the HT29 colon cancer-derived cell line leads to a reduced malignant phenotype. Here we show that restoring Cdx1 and Cdx2 expression in HT29 cells enhanced the antigen presentation system, as reflected by a strong induction of the concentration of HLA-I molecules at the cell surface, resulting from increased expression of the HLA-I mRNA. Expression of the LMP2 proteasomal protein was also strongly induced by Cdx1 and Cdx2 at the transcriptional level, whereas TAP1 expression which is under the control of the same bidirectional promoter as LMP2 remained unchanged. Furthermore, expression of the adhesion molecule ICAM-1, which works in concert with HLA-I, and of the cell death promoter Fas was also increased upon Cdx1 and Cdx2 expression. Taken together, these results suggest that loss of Cdx1 and Cdx2 expression during colorectal carcinogenesis could favor the escape of tumor cells from the immune system. In conclusion, restoration of Cdx1 and Cdx2 expression should be considered in immunotherapeutic strategies for colorectal cancer.
Collapse
Affiliation(s)
- P Soubeyran
- Laboratoire de Recherche de Physiologie et Pathologie Digestives, INSERM U.315, 46 Boulevard de la Gaye, Marseille, 13009, France
| | | | | | | | | |
Collapse
|
35
|
Ribas A, Ribas-Mundó M. [Antitumor immunology in the year 2000 and the new immunosuppressive therapy]. Med Clin (Barc) 2000; 114:579-83. [PMID: 10846678 DOI: 10.1016/s0025-7753(00)71369-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- A Ribas
- División de Hematología-Oncología, Universidad de California, Los Angeles, USA
| | | |
Collapse
|
36
|
Aidoo M, Udhayakumar V. Field studies of cytotoxic T lymphocytes in malaria infections: implications for malaria vaccine development. PARASITOLOGY TODAY (PERSONAL ED.) 2000; 16:50-6. [PMID: 10652487 DOI: 10.1016/s0169-4758(99)01592-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The search for a cytotoxic T lymphocyte (CTL)-inducing malaria vaccine has moved forward from epitope identification to planning stages of safety and immunogenicity trials of candidate vaccines. Development of CTL-inducing vaccine candidates has taken center stage based on the observation that CTL-mediated protection might be the dominant mechanism by which sterile immunity is achieved in irradiated sporozoite immunization experiments in humans and laboratory animals. However, studies in naturally infected individuals living in endemic areas, as reviewed here by Michael Aidoo and Venkatachalam Udhayakumar, have revealed that CTL induction might be influenced by factors such as parasite variants, host genes, other infections and transmission patterns. The influence of these factors on CTL induction has been demonstrated individually and in various combinations in controlled animal experiments. However, in naturally infected humans, they are presented in a complex host-parasite-environment interaction, in a manner that is not easily achieved in laboratory-based experiments. Understanding these interactions is crucial for the development and testing of CTL-inducing vaccines for humans.
Collapse
Affiliation(s)
- M Aidoo
- Molecular Vaccine Section, Immunology Branch, Division of Parasitic Diseases, National Center for Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30341, USA.
| | | |
Collapse
|
37
|
Leite-de-Moraes MC, Hameg A, Arnould A, Machavoine F, Koezuka Y, Schneider E, Herbelin A, Dy M. A Distinct IL-18-Induced Pathway to Fully Activate NK T Lymphocytes Independently from TCR Engagement. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.11.5871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
NK T lymphocytes are characterized by their ability to promptly generate IL-4 and IFN-γ upon TCR engagement. Here, we demonstrate that these cells can also be fully activated in the absence of TCR cross-linking in response to the proinflammatory cytokine IL-18 associated with IL-12. NK T cells stimulated with IL-18 plus IL-12 proliferated, killed Fas+ target cells, and produced high levels of IFN-γ without IL-4. In these conditions, IFN-γ production was at least 10-fold higher than that upon TCR cross-linking. Interestingly, a 2-h pretreatment with IL-12 plus IL-18 sufficed to maintain the high IFN-γ-producing potential during subsequent stimulation with anti-TCR mAbs or with the specific Ag α-galactosylceramide. Similar effects were observed in vivo, because splenic CD4+ NK T cells from MHC class II-deficient mice secreted IFN-γ without further stimulation when removed 2 h after a single injection of IL-12 plus IL-18. In conclusion, our evidence for activation of NK T lymphocytes in response to IL-18 plus IL-12 in the absence of TCR engagement together with the maintenance of preferential IFN-γ vs IL-4 production upon subsequent exposure to specific Ags is consistent with the active participation of this cell population in innate as well as acquired cellular immune responses.
Collapse
Affiliation(s)
- Maria C. Leite-de-Moraes
- *Centre National de la Recherche Scientifique, UMR 8603, Université René Descartes, Paris V, and
| | - Agathe Hameg
- †Institut National de la Santé et de la Recherche, Unité 25, Hôpital Necker, Paris, France; and
| | - Anne Arnould
- *Centre National de la Recherche Scientifique, UMR 8603, Université René Descartes, Paris V, and
| | - François Machavoine
- *Centre National de la Recherche Scientifique, UMR 8603, Université René Descartes, Paris V, and
| | - Yasuhiko Koezuka
- ‡Pharmaceutical Research Laboratory, Kirin Brewery Co., Ltd., Gunma, Japan
| | - Elke Schneider
- *Centre National de la Recherche Scientifique, UMR 8603, Université René Descartes, Paris V, and
| | - André Herbelin
- †Institut National de la Santé et de la Recherche, Unité 25, Hôpital Necker, Paris, France; and
| | - Michel Dy
- *Centre National de la Recherche Scientifique, UMR 8603, Université René Descartes, Paris V, and
| |
Collapse
|
38
|
Lilić M, Kulig K, Messaoudi I, Remus K, Janković M, Nikolić-Zugić J, Vukmanović S. CD8(+) T cell cytolytic activity independent of mitogen-activated protein kinase / extracellular regulatory kinase signaling (MAP kinase / ERK). Eur J Immunol 1999; 29:3971-7. [PMID: 10602006 DOI: 10.1002/(sici)1521-4141(199912)29:12<3971::aid-immu3971>3.0.co;2-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Cytotoxicity is a major effector function of CD8(+) T cells. Although mitogen-activated protein kinase (MAP kinase) / extracellular regulatory kinase (ERK) activity is indispensable for cytotoxic activity of most CD8(+) T cells a portion of CD8(+) T cells appears resistant to MEK inhibition as cytotoxicity of bulk cultures was partially preserved in the presence of a MEK inhibitor. We have also identified a long-term CD8(+) T cell line with unaltered cytolytic activity after prevention of ERK activation. Antigen-induced microtubule organizing center (MTOC) reorientation was not prevented in this CD8(+) cell line by MEK inhibition, in sharp contrast to the MTOC reorientation prevention in a CD8(+) T cell clone with MEK inhibition-sensitive cytolytic activity. These findings suggest that resistance of lysis to MEK inhibition may be due to a lack of ERK control over MTOC reorientation in some CD8(+) T cells. Thus, there appears to be a heterogeneity of ERK-regulated cytolytic activity in CD8(+) T cells, most likely resulting from a differential control of ERK over MTOC motility.
Collapse
Affiliation(s)
- M Lilić
- Michael Heidelberger Division of Immunology Department of Pathology and Kaplan Comprehensive Cancer Center, NYU Medical Center, New York, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Manjunath N, Shankar P, Stockton B, Dubey PD, Lieberman J, von Andrian UH. A transgenic mouse model to analyze CD8(+) effector T cell differentiation in vivo. Proc Natl Acad Sci U S A 1999; 96:13932-7. [PMID: 10570176 PMCID: PMC24168 DOI: 10.1073/pnas.96.24.13932] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Antigen-specific effector T cells are prerequisite to immune protection, but because of the lack of effector cell-specific markers, their generation and differentiation has been difficult to study. We report that effector cells are highly enriched in a T cell subset that can be specifically identified in transgenic (T-GFP) mice expressing green fluorescent protein (GFP) under control of the murine CD4 promoter and proximal enhancer. Consistent with previous studies of these transcriptional control elements, GFP was strongly and specifically expressed in nearly all resting and short-term activated CD4(+) and CD8(+) T cells. However, when T-GFP mice were challenged with vaccinia virus, allogeneic tumor cells, or staphylococcal enterotoxin A, the cytotoxic and IFN-gamma-producing T cells lost GFP expression. Upon T cell receptor (TCR) ligation by alphaCD3, sorted GFP(+) cells fluxed calcium and proliferated vigorously. In contrast, GFP(-) effector cells showed a diminished calcium flux and did not proliferate. Instead, they underwent apoptosis unless supplied with exogenous IL-2. By reverse transcription-PCR analysis, the GFP(-) cells up-regulated the pro-apoptotic molecule, Fas-L, and down-regulated gene expression of the proximal TCR signaling molecule, CD3zeta, and c-jun, a component of the AP-1 transcription factor. Thus, differential regulation of TCR signaling may explain the divergent responses of naïve and effector T cells to antigen stimulation.
Collapse
Affiliation(s)
- N Manjunath
- The Center for Blood Research, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
40
|
Kreuwel HTC, Morgan DJ, Krahl T, Ko A, Sarvetnick N, Sherman LA. Comparing the Relative Role of Perforin/Granzyme Versus Fas/Fas Ligand Cytotoxic Pathways in CD8+ T Cell-Mediated Insulin-Dependent Diabetes Mellitus. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.8.4335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
CD8+ cytotoxic T cells play a critical role in initiating insulin-dependent diabetes mellitus. The relative contribution of each of the major cytotoxic pathways, perforin/granzyme and Fas/Fas ligand (FasL), in the induction of autoimmune diabetes remains controversial. To evaluate the role of each lytic pathway in β cell lysis and induction of diabetes, we have used a transgenic mouse model in which β cells expressing the influenza virus hemagglutinin (HA) are destroyed by HA-specific CD8+ T cells from clone-4 TCR-transgenic mice. Upon adoptive transfer of CD8+ T cells from perforin-deficient clone-4 TCR mice, there was a 30-fold increase in the number of T cells required to induce diabetes. In contrast, elimination of the Fas/FasL pathway of cytotoxicity had little consequence. When both pathways of cytolysis were eliminated, mice did not become diabetic. Using a model of spontaneous diabetes, which occurs in double transgenic neonates that express both clone-4 TCR and Ins-HA transgenes, mice deficient in either the perforin or FasL/Fas lytic pathway become diabetic soon after birth. This indicates that, in the neonate, large numbers of autoreactive CD8+ T cells can lead to destruction of islet β cells by either pathway.
Collapse
Affiliation(s)
- Huub T. C. Kreuwel
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037
| | - David J. Morgan
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037
| | - Troy Krahl
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037
| | - Alice Ko
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037
| | - Nora Sarvetnick
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037
| | - Linda A. Sherman
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037
| |
Collapse
|
41
|
Sol MA, Vacaresse N, Lule J, Davrinche C, Gabriel B, Teissie J, Ziegler A, Thomsen M, Benoist H. N-linked oligosaccharides can protect target cells from the lysis mediated by NK cells but not by cytotoxic T lymphocytes: role of NKG2-A. TISSUE ANTIGENS 1999; 54:113-21. [PMID: 10488737 DOI: 10.1034/j.1399-0039.1999.540201.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We have previously shown that glycophorin A (GPA), inserted by electropulsation into the membrane of K562 cells, protected them from natural killer (NK) cell-mediated cytotoxicity and the unique N-linked oligosaccharide of GPA was essential for resistance to occur. The present study demonstrates that the protection level conferred by GPA is similar to the resistance induced by HLA-Cw3 expressed by transfected K562 cells. A monoclonal antibody against NKG2-A, an NK inhibitory receptor interacting with HLA class I antigens and belonging to the C-type lectin receptor, was able to restore the ability of NK cells to lyse K562 cells expressing HLA-Cw3 at the cell membrane but not electroinserted-GPA, suggesting that the N-linked oligosaccharide of GPA cannot be a ligand for NKG2-A. GPA was then electroinserted into the membrane of two lymphoblastoid B-cell lines: one was sensitive to NK cell-mediated lysis, the other was susceptible to cytotoxic CD8+ T-lymphocyte (CTL)-mediated cytotoxicity. The electroinserted GPA protected the target cells from NK-mediated cytotoxicity, whereas it did not modify the cell susceptibility to lysis by CTL. Endoglycosidase F treatment abolished the resistance towards NK cell-mediated lysis, suggesting that N-linked glycans could inhibit mechanisms used by NK cells to exert their cytotoxic function in agreement with our previous results.
Collapse
Affiliation(s)
- M A Sol
- INSERM U 466, Institut Louis Bugnard, CHU Ranguell, Toulouse, France
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Gullo CA, Esser MT, Fuller CL, Braciale VL. Generation of IL-2-Dependent Cytolytic T Lymphocytes (CTLs) with Altered TCR Responses Derived from Antigen-Dependent CTL Clones. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.11.6466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Ag-specific CD8+ CTL clones require TCR stimulation to respond to IL-2 for growth. Because IL-2 may be produced in the vicinity of CD8+ CTLs when Ag is limiting at the end of an immune response, we have examined the effect of culturing viral-specific CTL clones in IL-2 in the absence of antigenic stimulation. Limiting dilution analysis revealed a high precursor frequency for CTL clones derived from IL-2 propagation (termed CTL-factor dependent (FD)) that are dependent upon exogenous IL-2 for growth and survival and no longer require TCR stimulation to proliferate. Culturing CTL-FDs with infected splenocytes presenting Ag and IL-2 did not revert the clones but did lead to a TCR-induced inhibition of proliferation. The derived CTL-FDs have lost the ability to kill via the perforin/granule exocytosis mechanism of killing, although they express similar levels of TCR, CD3ε, CD8αβ, CD45, and LFA-1 compared with the parental clones. The CTL-FDs retain Fas ligand/Fas-mediated cytotoxicity, and IFN-γ production and regulate the expression of CD69 and IL-2Rα when triggered through the TCR. A parental CTL protected BALB/c mice from a lethal challenge of influenza virus, whereas a CTL-FD did not. These findings represent a novel regulatory function of IL-2 in vitro that, if functional in vivo, may serve to down-regulate cellular immune responses.
Collapse
Affiliation(s)
- Charles A. Gullo
- Department of Microbiology and Beirne B. Carter Center for Immunology Research, University of Virginia Health Sciences Center, Charlottesville, VA 22908
| | - Mark T. Esser
- Department of Microbiology and Beirne B. Carter Center for Immunology Research, University of Virginia Health Sciences Center, Charlottesville, VA 22908
| | - Claudette L. Fuller
- Department of Microbiology and Beirne B. Carter Center for Immunology Research, University of Virginia Health Sciences Center, Charlottesville, VA 22908
| | - Vivian Lam Braciale
- Department of Microbiology and Beirne B. Carter Center for Immunology Research, University of Virginia Health Sciences Center, Charlottesville, VA 22908
| |
Collapse
|
43
|
Fuller CL, Ravichandran KS, Braciale VL. Phosphatidylinositol 3-Kinase-Dependent and -Independent Cytolytic Effector Functions. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.11.6337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
Two distinct forms of short-term cytolysis have been described for CD8+ CTLs, the perforin/granzyme- and Fas ligand/Fas (CD95 ligand (CD95L)/CD95)-mediated pathways. However, the difference in signal transduction events leading to these cytolytic mechanisms remains unclear. We used wortmannin, an irreversible antagonist of phosphatidylinositol 3-kinase (PI3-K) activity, to investigate the role of PI3-K in influenza-specific CD8+ CTL cytolytic effector function. We found that the addition of wortmannin at concentrations as low as 1 nM significantly inhibited both the Ag/MHC-induced cytolysis of CD95− target cells and serine esterase release. In strong contrast, W did not inhibit the Ag/MHC-induced CD95L expression or the CD95L/CD95-mediated cytolysis of CD95+ targets. A combination of wortmannin and blocking mAb against CD95L inhibited the cytolysis of CD95+ targets, indicating that the wortmannin-independent cytolysis was due to CD95L/CD95 mediated cytolysis. These findings suggest a differential role for PI3-K in mediating cytolysis and, thus far, the earliest difference between perforin/granzyme- and CD95L/CD95-dependent cytolysis. Our data reinforce the idea of a TCR with modular signal transduction pathways that can be triggered or inhibited selectively, resulting in differential effector function.
Collapse
Affiliation(s)
- Claudette L. Fuller
- *Department of Microbiology,
- †Beirne B. Carter Center for Immunology Research, University of Virginia Health Sciences Center, Charlottesville, VA 22908; and
| | - Kodimangalam S. Ravichandran
- *Department of Microbiology,
- †Beirne B. Carter Center for Immunology Research, University of Virginia Health Sciences Center, Charlottesville, VA 22908; and
| | - Vivian L. Braciale
- *Department of Microbiology,
- †Beirne B. Carter Center for Immunology Research, University of Virginia Health Sciences Center, Charlottesville, VA 22908; and
- ‡Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555
| |
Collapse
|
44
|
Yasukawa M, Ohminami H, Yakushijin Y, Arai J, Hasegawa A, Ishida Y, Fujita S. Fas-Independent Cytotoxicity Mediated by Human CD4+ CTL Directed Against Herpes Simplex Virus-Infected Cells. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.10.6100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
The present study was undertaken to clarify the mechanisms of cytotoxicity mediated by virus-specific human CD4+ CTLs using the lymphocytes of family members with a Fas gene mutation. CD4+ CTL bulk lines and clones directed against HSV-infected cells were established from lymphocytes of a patient with a homozygous Fas gene mutation and of the patient’s mother. HSV-specific CD4+ CTLs generated from lymphocytes of the patient and her mother exerted cytotoxicity against HSV-infected cells from the patient (Fas−/−) and from her mother (Fas+/−) to almost the same degree in an HLA class II-restricted manner. mRNAs for the major mediators of CTL cytotoxicity, Fas ligand, perforin, and granzyme B, were detected in these CD4+ CTLs using the RT-PCR and flow cytometry. The cytotoxicity of the HSV-specific CD4+ CTLs appeared to be Ca2+-dependent and was almost completely inhibited by concanamycin A, a potent inhibitor of the perforin-based cytotoxic pathway. Although the Fas/Fas ligand system has been reported to be the most important mechanism for CD4+ CTL-mediated cytotoxicity in the murine system, the present findings strongly suggest that granule exocytosis, not the Fas/Fas ligand system, is the main pathway for the cytotoxicity mediated by HSV-specific human CD4+ CTLs.
Collapse
Affiliation(s)
| | | | | | - Junko Arai
- *First Department of Internal Medicine and
| | | | - Yasushi Ishida
- †Department of Pediatrics, Ehime University School of Medicine, Shigenobu, Ehime, Japan
| | | |
Collapse
|
45
|
Yi S, Feng X, Wang Y, Kay TW, Wang Y, O'Connell PJ. CD4+ cells play a major role in xenogeneic human anti-pig cytotoxicity through the Fas/Fas ligand lytic pathway. Transplantation 1999; 67:435-43. [PMID: 10030292 DOI: 10.1097/00007890-199902150-00017] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND In this study, the role of cell-mediated cytotoxicity by human leukocytes against pig endothelial cells was examined in vitro. The aim was to determine which cell subsets were responsible for this phenomenon and which pathways were involved in cell lysis. METHODS Primed human peripheral blood mononuclear cells (PBMC) or purified CD4+ or CD8+ T cells were used in a cell-mediated cytotoxicity assay in which cytotoxicity of an SV40 transformed porcine endothelial cell (EC) line (SVAP) was determined by Annexin V binding. RESULTS Human PBMC demonstrated specific lysis of porcine EC that was proportional to the effector: target ratio. CD4+ T cells accounted for >60% of this lysis, whereas CD8+ T cells accounted for <20%. CD4+ T cell-mediated lysis depended on direct recognition of porcine major histocompatibility complex class II molecules as inhibition of swine leukocyte antigen class II on porcine EC-inhibited CD4+ T cell cytotoxicity. This lysis was mediated through the Fas/FasL pathway as addition of anti-Fas and/or anti-FasL antibody profoundly inhibited antiporcine lysis. In addition, FasL gene expression was detected in primed PBMC and CD4+ T cells by RT-PCR, whereas granzyme B gene expression was not. Primed CD4+ T cells demonstrated high level FasL protein by Western blotting and two-color FACS analysis, whereas NK cells and CD8+ T cells did not. Finally, recombinant human FasL induced apoptosis in Fas expressing porcine EC cells, demonstrating that human FasL interacted with and activated Fas on porcine EC cells. CONCLUSIONS In conclusion, human to pig cell-mediated cytotoxicity was mediated predominantly by CD4+ T cells through the Fas/FasL pathway of apoptosis. These results suggest that direct cytotoxicity by xenoreactive CD4+ T cells may be one of several effector mechanisms involved in cellular xenograft rejection.
Collapse
Affiliation(s)
- S Yi
- National Pancreas Transplant Unit, Westmead Hospital, NSW, Australia
| | | | | | | | | | | |
Collapse
|
46
|
Kessler B, Hudrisier D, Schroeter M, Tschopp J, Cerottini JC, Luescher IF. Peptide Modification or Blocking of CD8, Resulting in Weak TCR Signaling, Can Activate CTL for Fas- But Not Perforin-Dependent Cytotoxicity or Cytokine Production. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.12.6939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Abstract
This study describes a form of partial agonism for a CD8+CTL clone, S15, in which perforin-dependent killing and IFN-γ production were lost but Fas (APO1 or CD95)-dependent cytotoxicity preserved. Cloned S15 CTL are H-2Kd restricted and specific for a photoreactive derivative of the Plasmodium berghei circumsporozoite peptide PbCS 252–260 (SYIPSAEKI). The presence of a photoactivatable group in the epitope permitted assessment of TCR-ligand binding by TCR photoaffinity labeling. Selective activation of Fas-dependent killing was observed for a peptide-derivative variant containing a modified photoreactive group. A similar functional response was obtained after binding of the wild-type peptide derivative upon blocking of CD8 participation in TCR-ligand binding. The epitope modification or blocking of CD8 resulted in an ≥8-fold decrease in TCR-ligand binding. In both cases, phosphorylation of ζ-chain and ZAP-70, as well as calcium mobilization were reduced close to background levels, indicating that activation of Fas-dependent cytotoxicity required weaker TCR signaling than activation of perforin-dependent killing or IFN-γ production. Consistent with this, we observed that depletion of the protein tyrosine kinase p56lck by preincubation of S15 CTL with herbimycin A severely impaired perforin- but not Fas-dependent cytotoxicity. Together with the observation that S15 CTL constitutively express Fas ligand, these results indicate that TCR signaling too weak to elicit perforin-dependent cytotoxicity or cytokine production can induce Fas-dependent cytotoxicity, possibly by translocation of preformed Fas ligand to the cell surface.
Collapse
Affiliation(s)
| | | | - Michael Schroeter
- †Institute of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Jürg Tschopp
- †Institute of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | | | | |
Collapse
|
47
|
Fuller CL, Braciale VL. Selective Induction of CD8+ Cytotoxic T Lymphocyte Effector Function by Staphylococcus Enterotoxin B. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.10.5179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
Upon encounter with its antigenic stimulus, CTL characteristically proliferate, produce cytokines, and lyse the Ag-presenting cell in an attempt to impede further infection. Superantigens are extremely efficient immunostimulatory proteins that promote high levels of proliferation and massive cytokine production in reactive T cells. We compared the activation of murine influenza-specific CD8+ CTL clones stimulated with either influenza peptide or the superantigen staphylococcus enterotoxin B (SEB). We found that influenza peptide/MHC and SEB appeared equally capable of eliciting proliferation and IFN-γ production. However, while influenza peptide/MHC elicited both perforin- and Fas ligand (FasL)/Fas (CD95L/CD95)-mediated cytolytic mechanisms, SEB was unable to trigger perforin-mediated cytolysis or serine esterase release. Examination of intracellular Ca2+ mobilization events revealed that the ability to trigger intracellular Ca2+ flux was not comparable between influenza peptide and SEB. SEB stimulated only a small rise in levels of intracellular Ca2+, at times indistinguishable from background. These findings indicate that the short-term cytolytic potential of superantigen-activated CD8+ CTL clones appears to be restricted to FasL/Fas (CD95L/CD95) mediated cytolysis.
Collapse
Affiliation(s)
- Claudette L. Fuller
- Department of Microbiology and Beirne B. Carter Center for Immunology Research, Health Sciences Center, University of Virginia, Charlottesville, VA 22908
| | - Vivian Lam Braciale
- Department of Microbiology and Beirne B. Carter Center for Immunology Research, Health Sciences Center, University of Virginia, Charlottesville, VA 22908
| |
Collapse
|
48
|
Rivoltini L, Radrizzani M, Accornero P, Squarcina P, Chiodoni C, Mazzocchi A, Castelli C, Tarsini P, Viggiano V, Belli F, Colombo MP, Parmiani G. Human Melanoma-Reactive CD4+ and CD8+ CTL Clones Resist Fas Ligand-Induced Apoptosis and Use Fas/Fas Ligand-Independent Mechanisms for Tumor Killing. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.3.1220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
Tumor cells have been shown recently to escape immune recognition by developing resistance to Fas-mediated apoptosis and acquiring expression of Fas ligand (FasL) molecule that they may use for eliminating activated Fas+ lymphocytes. In this study, we report that tumor-specific T lymphocytes isolated from tumor lesions by repeated in vitro TCR stimulation with relevant Ags (mostly represented by normal self proteins, such as MART-1/Melan A and gp100) can develop strategies for overcoming these escape mechanisms. Melanoma cells (and normal melanocytes) express heterogeneous levels of Fas molecule, but they result homogeneously resistant to Fas-induced apoptosis. However, CD4+ and CD8+ CTL clones kill melanoma cells through Fas/FasL-independent, granule-dependent lytic pathway. In these lymphocytes, Ag/MHC complex interaction with TCR does not lead to functional involvement of FasL, triggered, on the contrary, by T cell activation with nonspecific stimuli such as PMA/ionomycin. Additionally, melanoma cells express significant levels of FasL (detectable on the cell surface only after treatment with metalloprotease inhibitors), although to a lesser extent than professional immune cells such as Th1 clones. Nevertheless, antimelanoma CTL clones resist apoptosis mediated by FasL either in soluble form or expressed by Th1 lymphocytes or FasL+ melanoma cells. These results demonstrate that CD4+ and CD8+ antimelanoma T cell clones can be protected against Fas-dependent apoptosis, and thus be useful reagents of immunotherapeutic strategies aimed to potentiate tumor-specific T cell responses.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Filiberto Belli
- ‡Surgical Oncology B, Istituto Nazionale Tumori, Milan, Italy
- Divisions of
| | | | | |
Collapse
|
49
|
Zeine R, Pon R, Ladiwala U, Antel JP, Filion LG, Freedman MS. Mechanism of gammadelta T cell-induced human oligodendrocyte cytotoxicity: relevance to multiple sclerosis. J Neuroimmunol 1998; 87:49-61. [PMID: 9670845 DOI: 10.1016/s0165-5728(98)00047-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Gammadelta T cells may contribute to the pathogenesis of Multiple Sclerosis (MS) via cytotoxicity directed at the myelin-oligodendrocyte unit. We have previously demonstrated that peripheral blood-derived gammadelta T cells lyse fresh human oligodendrocytes in vitro. The present work extends these observations to gammadelta T cells derived from both peripheral blood (PBL) and cerebrospinal fluid (CSF) of MS and non-MS neurological disease controls and addresses the mechanism of cellular cytotoxicity. We found that MS patients contained increased proportions of Vdelta1+ gammadelta T cells in both CSF and PBL samples compared to other neurological disease (OND) controls. Although gammadelta T cells from all patients were cytotoxic towards Daudi, RPMI 8226, U937, Jurkat, oligodendroglioma and fresh human oligodendrocyte targets, OND-derived, Vdelta2+ rich, populations derived from the CSF exhibited greater cytotoxicity towards cell lines (Daudi, RPMI 8226) known to express high levels of heat shock proteins (hsp). To clarify the mechanism(s) of cytotoxicity used by gammadelta T cells, we first showed that cell-target contact was necessary by the use of physical barriers (transwells), which reduced target cell lysis by at least 75%. The use of Ca2+-free media reduced lysis by up to 50%, but fully blocking gammadelta T cell Perforin release and function by either Ca2+ chelation (Mg2EGTA) or the H+-ATPase inhibitor Concanamycin-A (CMA), completely abrogated the lysis of Fas-/hsp60high expressing targets (Daudi, U937). However, additional treatment with Brefeldin A was required for the complete inhibition of gammadelta T cell mediated killing of Fas+ expressing Jurkat targets and fresh human brain-derived oligodendrocytes. Inhibition of granzyme activity by an isocoumarin compound reduced cytolysis only slightly. The use of either Brefeldin A or an anti-Fas antibody alone did not significantly affect lysis. These findings suggest that in MS, gammadelta T cells may utilize either the Fas-mediated or Perforin-based cell cytotoxicity pathways in exerting oligodendrocyte damage, though the Perforin pathway is predominant.
Collapse
Affiliation(s)
- R Zeine
- Department of Medicine, University of Ottawa, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
50
|
Tsujimura K, Takahashi T, Iwase S, Matsudaira Y, Kaneko Y, Yagita H, Obata Y. Two Types of Anti-TL (Thymus Leukemia) CTL Clones with Distinct Target Specificities: Differences in Cytotoxic Mechanisms and Accessory Molecule Requirements. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.160.11.5253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
TCRαβ CTL clones recognizing mouse thymus leukemia (TL) Ags were established and categorized into two groups: those killing any TL+ target cells (type I) and those killing only TL+ Con A blasts (type II). Cold target inhibition assays showed that the antigenic determinant(s) recognized by type II clones are expressed not only on TL+ Con A blasts but also on other TL+ target cells. The relation of the target specificity to the killing machinery and the accessory molecules involved in cytotoxicity were therefore analyzed using four representative clones selected from each type. Of the target cells tested, Fas was only expressed on Con A blasts, indicating that Fas ligand (FasL)-dependent cytotoxicity is limited to such cells. All four type II and one of four type I clones expressed FasL on the surface, while both types contained perforin in the cytoplasm. Blocking studies using neutralizing anti-FasL mAbs and concanamycin A (CMA), a selective inhibitor of the perforin pathway, suggested that type I clones kill target cells by way of perforin, while type II clones kill TL+ Con A blasts through FasL together with perforin. For their cytotoxicity, type I CTLs require a signal through CD8, while type II require LFA-1/ICAM-1 interactions. Type II clones also need a costimulatory signal through an unknown molecule for perforin-dependent cytotoxicity. These results taken together suggest that the difference in the target specificity of anti-TL CTL clones is due to variation in the killing machineries and the dependence on accessory molecules.
Collapse
Affiliation(s)
- Kunio Tsujimura
- *Laboratory of Immunology, Aichi Cancer Center Research Institute, Nagoya
| | | | - Shigeru Iwase
- *Laboratory of Immunology, Aichi Cancer Center Research Institute, Nagoya
- †Department of Chemical Hygiene and Nutrition, Faculty of Pharmaceutical Sciences, Nagoya City University, Nagoya
| | - Yasue Matsudaira
- *Laboratory of Immunology, Aichi Cancer Center Research Institute, Nagoya
| | - Yoko Kaneko
- ‡Department for Geriatric Research, National Institute for Longevity Sciences, Obu; and
| | - Hideo Yagita
- §Department of Immunology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yuichi Obata
- *Laboratory of Immunology, Aichi Cancer Center Research Institute, Nagoya
| |
Collapse
|