51
|
Weist BJD, Wehler P, El Ahmad L, Schmueck-Henneresse M, Millward JM, Nienen M, Neumann AU, Reinke P, Babel N. A revised strategy for monitoring BKV-specific cellular immunity in kidney transplant patients. Kidney Int 2015. [PMID: 26221751 DOI: 10.1038/ki.2015.215] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Reactivation of Polyomavirus BKV is a severe complication in kidney transplant patients. Current treatment requires close monitoring, and modification of immunosuppressive drugs. As an important additional tool, the monitoring of BKV immunity has been based on detection of cytokine-secreting T cells upon BKV-antigen challenge. However, low frequent BKV-specific T cells are often barely detectable and their roles in BKV clearance remain unclear. Here, we analyzed the effects of immunosuppressive agents on BKV-specific T cells in vitro. Significant reductions in expression of several markers, and reduced killing functions upon treatment with calcineurin but not mTOR inhibitors were detected. However, effects of these drugs on expression of surface markers and GranzymeB were substantially less striking than effects on cytokine expression. Consequently, we applied a novel detection strategy for BKV-specific T cells in immunosuppressed kidney transplant patients using these more robust markers, and showed significantly improved sensitivity compared with the conventional IFNγ-based method. Using this strategy and 17-color flow cytometry, we found BKV-specific helper and cytolytic CD4+ T-cell subsets that differed in their memory phenotype, which corresponded with BKV clearance in kidney transplant patients. Thus, our results offer an improved detection strategy for BKV-specific T cells in kidney transplant patients, and shed light on the contributions of these cells to BKV clearance.
Collapse
Affiliation(s)
- Benjamin J D Weist
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Germany
| | - Patrizia Wehler
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Germany
| | - Linda El Ahmad
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Germany
| | | | - Jason M Millward
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Germany
| | - Mikalai Nienen
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Germany
| | - Avidan U Neumann
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Germany
| | - Petra Reinke
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Germany.,Department of Nephrology, Charité Universitätsmedizin Berlin, Germany
| | - Nina Babel
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Germany.,Medical Clinic I, Marien Hospital Herne, Ruhr University Bochum, Herne, Germany
| |
Collapse
|
52
|
Flacher V, Tripp CH, Mairhofer DG, Steinman RM, Stoitzner P, Idoyaga J, Romani N. Murine Langerin+ dermal dendritic cells prime CD8+ T cells while Langerhans cells induce cross-tolerance. EMBO Mol Med 2015; 6:1191-204. [PMID: 25085878 PMCID: PMC4197865 DOI: 10.15252/emmm.201303283] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Skin dendritic cells (DCs) control the immunogenicity of cutaneously administered vaccines. Antigens targeted to DCs via the C-type lectin Langerin/CD207 are cross-presented to CD8+ T cells in vivo. We investigated the relative roles of Langerhans cells (LCs) and Langerin+ dermal DCs (dDCs) in different vaccination settings. Poly(I:C) and anti-CD40 agonist antibody promoted cytotoxic responses upon intradermal immunization with ovalbumin (OVA)-coupled anti-Langerin antibodies (Langerin/OVA). This correlated with CD70 upregulation in Langerin+ dDCs, but not LCs. In chimeric mice where Langerin targeting was restricted to dDCs, CD8+ T-cell memory was enhanced. Conversely, providing Langerin/OVA exclusively to LCs failed to prime cytotoxicity, despite initial antigen cross-presentation to CD8+ T cells. Langerin/OVA combined with imiquimod could not prime CD8+ T cells and resulted in poor cytotoxicity in subsequent responses. This tolerance induction required targeting and maturation of LCs. Altogether, Langerin+ dDCs prime long-lasting cytotoxic responses, while cross-presentation by LCs negatively influences CD8+ T-cell priming. Moreover, this highlights that DCs exposed to TLR agonists can still induce tolerance and supports the existence of qualitatively different DC maturation programs.
Collapse
Affiliation(s)
- Vincent Flacher
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria Oncotyrol Center for Personalized Cancer Medicine, Innsbruck, Austria
| | - Christoph H Tripp
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria Oncotyrol Center for Personalized Cancer Medicine, Innsbruck, Austria
| | - David G Mairhofer
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria
| | - Ralph M Steinman
- Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, New York, NY, USA
| | - Patrizia Stoitzner
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria
| | - Juliana Idoyaga
- Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, New York, NY, USA
| | - Nikolaus Romani
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria Oncotyrol Center for Personalized Cancer Medicine, Innsbruck, Austria
| |
Collapse
|
53
|
Yusuf N, Nasti TH, Ahmad I, Chowdhury S, Mohiuddin H, Xu H, Athar M, Timares L, Elmets CA. In Vivo Suppression of Heat Shock Protein (HSP)27 and HSP70 Accelerates DMBA-Induced Skin Carcinogenesis by Inducing Antigenic Unresponsiveness to the Initiating Carcinogenic Chemical. THE JOURNAL OF IMMUNOLOGY 2015; 194:4796-803. [PMID: 25840912 DOI: 10.4049/jimmunol.1402804] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 03/13/2015] [Indexed: 01/07/2023]
Abstract
Heat shock proteins (HSPs) are constitutively expressed in murine skin. HSP27 is present in the epidermis, and HSP70 can be found in both the epidermis and dermis. The purpose of this study was to investigate the role of these proteins in cutaneous chemical carcinogenesis and to determine whether their effects on cell-mediated immune function were a contributing factor. In vivo inhibition of HSP27 and HSP70 produced a reduction in the T cell-mediated immune response to 7,12-dimethylbenz(a)anthracene (DMBA) and benzo(a)pyrene in C3H/HeN mice and resulted in a state of Ag-specific tolerance. When mice were pretreated with anti-HSP27 and anti-HSP70 Abs in vivo prior to subjecting them to a standard two-stage DMBA/12-O-tetradecanoylphorbol-13-acetate cutaneous carcinogenesis protocol, the percentage of mice with tumors was much greater (p < 0.05) in anti-HSP27- and HSP70-pretreated animals compared with mice pretreated with control Ab. Similar results were obtained when the data were evaluated as the cumulative number of tumors per group. Mice pretreated with HSP27 and HSP70 Abs developed more H-ras mutations and fewer DMBA-specific cytotoxic T lymphocytes. These findings indicate that in mice HSP27 and HSP70 play a key role in the induction of cell-mediated immunity to carcinogenic polyaromatic hydrocarbons. Bolstering the immune response to carcinogenic polyaromatic hydrocarbons may be an effective method for prevention of the tumors that they produce.
Collapse
Affiliation(s)
- Nabiha Yusuf
- Department of Dermatology and Skin Diseases Research Center, University of Alabama, Birmingham, AL 35294; and Veteran Affairs Medical Center, Birmingham, AL 35294
| | - Tahseen H Nasti
- Department of Dermatology and Skin Diseases Research Center, University of Alabama, Birmingham, AL 35294; and
| | - Israr Ahmad
- Department of Dermatology and Skin Diseases Research Center, University of Alabama, Birmingham, AL 35294; and
| | - Sanim Chowdhury
- Department of Dermatology and Skin Diseases Research Center, University of Alabama, Birmingham, AL 35294; and
| | - Hasan Mohiuddin
- Department of Dermatology and Skin Diseases Research Center, University of Alabama, Birmingham, AL 35294; and
| | - Hui Xu
- Department of Dermatology and Skin Diseases Research Center, University of Alabama, Birmingham, AL 35294; and Veteran Affairs Medical Center, Birmingham, AL 35294
| | - Mohammad Athar
- Department of Dermatology and Skin Diseases Research Center, University of Alabama, Birmingham, AL 35294; and
| | - Laura Timares
- Department of Dermatology and Skin Diseases Research Center, University of Alabama, Birmingham, AL 35294; and Veteran Affairs Medical Center, Birmingham, AL 35294
| | - Craig A Elmets
- Department of Dermatology and Skin Diseases Research Center, University of Alabama, Birmingham, AL 35294; and Veteran Affairs Medical Center, Birmingham, AL 35294
| |
Collapse
|
54
|
Zhang W, Oda T, Yu Q, Jin JO. Fucoidan from Macrocystis pyrifera has powerful immune-modulatory effects compared to three other fucoidans. Mar Drugs 2015; 13:1084-104. [PMID: 25706632 PMCID: PMC4377974 DOI: 10.3390/md13031084] [Citation(s) in RCA: 131] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 02/02/2015] [Accepted: 02/06/2015] [Indexed: 12/22/2022] Open
Abstract
Fucoidan, a sulfated polysaccharide purified from brown algae, has a variety of immune-modulation effects, such as promoting activation of dendritic cells (DCs), natural killer (NK) cells and T cells, and enhancing anti-viral and anti-tumor responses. However, the immune-modulatory effect of fucoidan from different seaweed extracts has not been thoroughly analyzed and compared. We analyzed fucoidans obtained from Ascophyllum nodosum (A. nodosum), Macrocystis pyrifera (M. pyrifera), Undaria pinnatifida (U. pinnatifida) and Fucus vesiculosus (F. vesiculosus) for their effect on the apoptosis of human neutrophils, activation of mouse NK cells, maturation of spleen DCs, proliferation and activation of T cells, and the adjuvant effect in vivo. Fucoidans from M. pyrifera and U. pinnatifida strongly delayed human neutrophil apoptosis at low concentration, whereas fucoidans from A. nodosum and F. vesiculosus delayed human neutrophil apoptosis at higher concentration. Moreover, fucoidan from M. pyrifera promoted NK cell activation and cytotoxic activity against YAC-1 cells. In addition, M. pyrifera fucoidan induced the strongest activation of spleen DCs and T cells and ovalbumin (OVA) specific immune responses compared to other fucoidans. These data suggest that fucoidan from M. pyrifera can be potentially useful as a therapeutic agent for infectious diseases, cancer and an effective adjuvant for vaccine.
Collapse
Affiliation(s)
- Wei Zhang
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai 201508, China.
| | - Tatsuya Oda
- Division of Biochemistry, Faculty of Fisheries, Nagasaki University, 1-14 Bunkyo-machi, Nagasaki, Nagasaki 852-8521, Japan.
| | - Qing Yu
- Department of Immunology and Infectious Diseases, The Forsyth Institute, 245 First Street, Cambridge, MA 02142, USA.
| | - Jun-O Jin
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai 201508, China.
| |
Collapse
|
55
|
Dawoodji A, Chen JL, Shepherd D, Dalin F, Tarlton A, Alimohammadi M, Penna-Martinez M, Meyer G, Mitchell AL, Gan EH, Bratland E, Bensing S, Husebye ES, Pearce SH, Badenhoop K, Kämpe O, Cerundolo V. High frequency of cytolytic 21-hydroxylase-specific CD8+ T cells in autoimmune Addison's disease patients. THE JOURNAL OF IMMUNOLOGY 2014; 193:2118-26. [PMID: 25063864 DOI: 10.4049/jimmunol.1400056] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The mechanisms behind destruction of the adrenal glands in autoimmune Addison's disease remain unclear. Autoantibodies against steroid 21-hydroxylase, an intracellular key enzyme of the adrenal cortex, are found in >90% of patients, but these autoantibodies are not thought to mediate the disease. In this article, we demonstrate highly frequent 21-hydroxylase-specific T cells detectable in 20 patients with Addison's disease. Using overlapping 18-aa peptides spanning the full length of 21-hydroxylase, we identified immunodominant CD8(+) and CD4(+) T cell responses in a large proportion of Addison's patients both ex vivo and after in vitro culture of PBLs ≤20 y after diagnosis. In a large proportion of patients, CD8(+) and CD4(+) 21-hydroxylase-specific T cells were very abundant and detectable in ex vivo assays. HLA class I tetramer-guided isolation of 21-hydroxylase-specific CD8(+) T cells showed their ability to lyse 21-hydroxylase-positive target cells, consistent with a potential mechanism for disease pathogenesis. These data indicate that strong CTL responses to 21-hydroxylase often occur in vivo, and that reactive CTLs have substantial proliferative and cytolytic potential. These results have implications for earlier diagnosis of adrenal failure and ultimately a potential target for therapeutic intervention and induction of immunity against adrenal cortex cancer.
Collapse
Affiliation(s)
- Amina Dawoodji
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, United Kingdom
| | - Ji-Li Chen
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, United Kingdom
| | - Dawn Shepherd
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, United Kingdom
| | - Frida Dalin
- Centre of Molecular Medicine, Department of Medicine (Solna), Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Andrea Tarlton
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, United Kingdom
| | - Mohammad Alimohammadi
- Centre of Molecular Medicine, Department of Medicine (Solna), Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Marissa Penna-Martinez
- Division of Endocrinology, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany
| | - Gesine Meyer
- Division of Endocrinology, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany
| | - Anna L Mitchell
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, United Kingdom
| | - Earn H Gan
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, United Kingdom
| | - Eirik Bratland
- Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway; Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
| | - Sophie Bensing
- Centre of Molecular Medicine, Department of Medicine (Solna), Karolinska Institutet, 171 76 Stockholm, Sweden; Department of Molecular Medicine and Surgery, Karolinska Institutet, SE-171 77 Stockholm, Sweden; and
| | - Eystein S Husebye
- Department of Clinical Science, University of Bergen, N-5021 Bergen, Norway; Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
| | - Simon H Pearce
- Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne NE1 3BZ, United Kingdom
| | - Klaus Badenhoop
- Division of Endocrinology, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany
| | - Olle Kämpe
- Centre of Molecular Medicine, Department of Medicine (Solna), Karolinska Institutet, 171 76 Stockholm, Sweden; Science for Life Laboratory, Uppsala University 750 03, Uppsala, Sweden
| | - Vincenzo Cerundolo
- Medical Research Council Human Immunology Unit, Medical Research Council Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, United Kingdom;
| |
Collapse
|
56
|
Weist BJD, Schmueck M, Fuehrer H, Sattler A, Reinke P, Babel N. The role of CD4(+) T cells in BKV-specific T cell immunity. Med Microbiol Immunol 2014; 203:395-408. [PMID: 25052009 DOI: 10.1007/s00430-014-0348-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 07/05/2014] [Indexed: 12/11/2022]
Abstract
Reactivation of polyomavirus BK (BKV) infection represents a severe complication in kidney transplant (KTX) patients. We previously reported an association between a declining BK viral load and the reconstitution of CD4(+) T cell BKV-specific immunity in patients following kidney transplantation. However, the specific contribution of CD4(+) T cells in the regulation of BKV-replication is unknown. Nevertheless, in vitro enrichment of BKV-specific T cells and subsequent adoptive T cell transfer may improve the restoration of immune competence in KTX patients with BKV infection. To date, strategies to capture human BKV-specific T cells with the ensuing expansion to clinically useful numbers are lacking. Here, we demonstrated a comprehensive flow cytometric analysis of the BKV-specific T cell response that permits access to the majority of T cells specific for immunodominant BKV antigens. A full-spectrum evaluation of the BKV-specific T cell response was performed by stimulating peripheral blood mononuclear cells (PBMC) with a mixture of BKV immunodominant peptide pools at varying concentrations and measuring activation marker expression and cytokine secretion. We also examined the effects of co-stimulation and PBMC resting time prior to activation. We defined the narrow range of stimulation conditions that permit the capture and expansion of functional BKV-specific T cell lines. The generated BKV-specific T cell lines showed the highest specificity and functionality when the T cells were captured according to IFNγ-secretion. This study highlights the multifunctional and cytolytic BKV-specific CD4(+) T cells as a dominant population within the generated T cell product. This method offers a novel approach for the generation of BKV-specific T cell lines for adoptive immunotherapy and underscores the critical role of CD4(+) T cells in the clearance of BKV.
Collapse
Affiliation(s)
- B J D Weist
- Department of Nephrology, Charité University Medicine, Berlin, Germany
| | | | | | | | | | | |
Collapse
|
57
|
Quah BJC, Wijesundara DK, Ranasinghe C, Parish CR. The use of fluorescent target arrays for assessment of T cell responses in vivo. J Vis Exp 2014:e51627. [PMID: 24998253 DOI: 10.3791/51627] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
The ability to monitor T cell responses in vivo is important for the development of our understanding of the immune response and the design of immunotherapies. Here we describe the use of fluorescent target array (FTA) technology, which utilizes vital dyes such as carboxyfluorescein succinimidyl ester (CFSE), violet laser excitable dyes (CellTrace Violet: CTV) and red laser excitable dyes (Cell Proliferation Dye eFluor 670: CPD) to combinatorially label mouse lymphocytes into > 250 discernable fluorescent cell clusters. Cell clusters within these FTAs can be pulsed with major histocompatibility (MHC) class-I and MHC class-II binding peptides and thereby act as target cells for CD8(+) and CD4(+) T cells, respectively. These FTA cells remain viable and fully functional, and can therefore be administered into mice to allow assessment of CD8(+) T cell-mediated killing of FTA target cells and CD4(+) T cell-meditated help of FTA B cell target cells in real time in vivo by flow cytometry. Since > 250 target cells can be assessed at once, the technique allows the monitoring of T cell responses against several antigen epitopes at several concentrations and in multiple replicates. As such, the technique can measure T cell responses at both a quantitative (e.g. the cumulative magnitude of the response) and a qualitative (e.g. functional avidity and epitope-cross reactivity of the response) level. Herein, we describe how these FTAs are constructed and give an example of how they can be applied to assess T cell responses induced by a recombinant pox virus vaccine.
Collapse
Affiliation(s)
- Benjamin J C Quah
- Department of Immunology, John Curtin School of Medical Research, Australian National University;
| | - Danushka K Wijesundara
- Department of Immunology, John Curtin School of Medical Research, Australian National University
| | - Charani Ranasinghe
- Department of Immunology, John Curtin School of Medical Research, Australian National University
| | - Christopher R Parish
- Department of Immunology, John Curtin School of Medical Research, Australian National University
| |
Collapse
|
58
|
Jin JO, Zhang W, Du JY, Wong KW, Oda T, Yu Q. Fucoidan can function as an adjuvant in vivo to enhance dendritic cell maturation and function and promote antigen-specific T cell immune responses. PLoS One 2014; 9:e99396. [PMID: 24911024 PMCID: PMC4049775 DOI: 10.1371/journal.pone.0099396] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 05/14/2014] [Indexed: 12/21/2022] Open
Abstract
Fucoidan, a sulfated polysaccharide purified from brown algae, has a variety of immune-modulation effects, including promoting antigen uptake and enhancing anti-viral and anti-tumor effects. However, the effect of fucoidan in vivo, especially its adjuvant effect on in vivo anti-tumor immune responses, was not fully investigated. In this study, we investigated the effect of fucoidan on the function of spleen dendritic cells (DCs) and its adjuvant effect in vivo. Systemic administration of fucoidan induced up-regulation of CD40, CD80 and CD86 expression and production of IL-6, IL-12 and TNF-α in spleen cDCs. Fucoidan also promoted the generation of IFN-γ-producing Th1 and Tc1 cells in an IL-12-dependent manner. When used as an adjuvant in vivo with ovalbumin (OVA) antigen, fucoidan promoted OVA-specific antibody production and primed IFN-γ production in OVA-specific T cells. Moreover, fucoidan enhanced OVA-induced up-regulation of MHC class I and II on spleen cDCs and strongly prompted the proliferation of OVA-specific CD4 and CD8 T cells. Finally, OVA immunization with fucoidan as adjuvant protected mice from the challenge with B16-OVA tumor cells. Taken together, these results suggest that fucoidan can function as an adjuvant to induce Th1 immune response and CTL activation, which may be useful in tumor vaccine development.
Collapse
Affiliation(s)
- Jun-O Jin
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
- * E-mail:
| | - Wei Zhang
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiang-Yuan Du
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ka-Wing Wong
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tatsuya Oda
- Graduate School of Science and Technology, Nagasaki University, Nagasaki, Japan
| | - Qing Yu
- Department of Immunology and Infectious Diseases, The Forsyth Institute, Cambridge, Massachusetts, United States of America
| |
Collapse
|
59
|
Crosby EJ, Goldschmidt MH, Wherry EJ, Scott P. Engagement of NKG2D on bystander memory CD8 T cells promotes increased immunopathology following Leishmania major infection. PLoS Pathog 2014; 10:e1003970. [PMID: 24586170 PMCID: PMC3937277 DOI: 10.1371/journal.ppat.1003970] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 01/17/2014] [Indexed: 02/07/2023] Open
Abstract
One of the hallmarks of adaptive immunity is the development of a long-term pathogen specific memory response. While persistent memory T cells certainly impact the immune response during a secondary challenge, their role in unrelated infections is less clear. To address this issue, we utilized lymphocytic choriomeningitis virus (LCMV) and Listeria monocytogenes immune mice to investigate whether bystander memory T cells influence Leishmania major infection. Despite similar parasite burdens, LCMV and Listeria immune mice exhibited a significant increase in leishmanial lesion size compared to mice infected with L. major alone. This increased lesion size was due to a severe inflammatory response, consisting not only of monocytes and neutrophils, but also significantly more CD8 T cells. Many of the CD8 T cells were LCMV specific and expressed gzmB and NKG2D, but unexpectedly expressed very little IFN-γ. Moreover, if CD8 T cells were depleted in LCMV immune mice prior to challenge with L. major, the increase in lesion size was lost. Strikingly, treating with NKG2D blocking antibodies abrogated the increased immunopathology observed in LCMV immune mice, showing that NKG2D engagement on LCMV specific memory CD8 T cells was required for the observed phenotype. These results indicate that bystander memory CD8 T cells can participate in an unrelated immune response and induce immunopathology through an NKG2D dependent mechanism without providing increased protection. Cutaneous leishmaniasis has a wide spectrum of clinical presentations, from mild self-healing lesions to severe chronic infections. Differences in each individual's response are related to pathogen dose and the genetic and physiological status of the host, but exactly what causes the broad spectrum of disease is not well understood. Here we show that previous infection with a viral or bacterial pathogen led to increased immunopathology associated with L. major infection. This increase in immunopathology was not associated with any changes in parasite control and was characterized by an exaggerated inflammatory infiltrate into the site of infection. Ultimately, this increase in immunopathology was dependent on the presence of memory CD8 T cells from the previous infection and their expression of the NK cell receptor NKG2D, as depletion of these cells prior to infection with L. major or blockade of this receptor during infection ameliorated the disease. Our work suggests that the immunological history of a patient may be playing an underlying role in the pathology associated with leishmania infection and could be an important consideration for the understanding and treatment of this and other human diseases. This work also identifies the NKG2D pathway as a potential new target for therapeutic intervention.
Collapse
Affiliation(s)
- Erika J. Crosby
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Michael H. Goldschmidt
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - E. John Wherry
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Phillip Scott
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
60
|
Polysaccharides from Ganoderma formosanum function as a Th1 adjuvant and stimulate cytotoxic T cell response in vivo. Vaccine 2014; 32:401-8. [DOI: 10.1016/j.vaccine.2013.11.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2013] [Revised: 09/20/2013] [Accepted: 11/06/2013] [Indexed: 12/21/2022]
|
61
|
Perret R, Sierro SR, Botelho NK, Corgnac S, Donda A, Romero P. Adjuvants that improve the ratio of antigen-specific effector to regulatory T cells enhance tumor immunity. Cancer Res 2013; 73:6597-608. [PMID: 24048821 DOI: 10.1158/0008-5472.can-13-0875] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Antitumor immunity is strongly influenced by the balance of tumor antigen-specific effector T cells (Teff) and regulatory T cells (Treg). However, the impact that vaccine adjuvants have in regulating the balance of antigen-specific T-cell populations is not well understood. We found that antigen-specific Tregs were induced following subcutaneous vaccination with either OVA or melanoma-derived peptides, with a restricted expansion of Teffs. Addition of the adjuvants CpG-ODN or Poly(I:C) preferentially amplified Teffs over Tregs, dramatically increasing the antigen-specific Teff:Treg ratios and inducing polyfunctional effector cells. In contrast, two other adjuvants, imiquimod and Quil A saponin, favored an expansion of antigen-specific Tregs and failed to increase Teff:Treg ratios. Following therapeutic vaccination of tumor-bearing mice, high ratios of tumor-specific Teffs:Tregs in draining lymph nodes were associated with enhanced CD8(+) T-cell infiltration at the tumor site and a durable rejection of tumors. Vaccine formulations of peptide+CpG-ODN or Poly(I:C) induced selective production of proinflammatory type I cytokines early after vaccination. This environment promoted CD8(+) and CD4(+) Teff expansion over that of antigen-specific Tregs, tipping the Teff to Treg balance to favor effector cells. Our findings advance understanding of the influence of different adjuvants on T-cell populations, facilitating the rational design of more effective cancer vaccines.
Collapse
Affiliation(s)
- Rachel Perret
- Authors' Affiliation: Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
62
|
Imai T, Ishida H, Suzue K, Hirai M, Taniguchi T, Okada H, Suzuki T, Shimokawa C, Hisaeda H. CD8(+) T cell activation by murine erythroblasts infected with malaria parasites. Sci Rep 2013; 3:1572. [PMID: 23535896 PMCID: PMC3610137 DOI: 10.1038/srep01572] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 03/11/2013] [Indexed: 01/30/2023] Open
Abstract
Recent studies show that some human malaria parasite species Plasmodium falciparum and P.vivax parasitize erythroblasts; however, the biological and clinical significance of this is unclear. To investigate further, we generated a rodent malaria parasite (P. yoelii 17XNL) expressing GFP-ovalbumin (OVA). Its infectivity to erythroblasts was confirmed, and parasitized erythroblasts were capable of initiating malaria infections. Experiments showed that MHC class I molecules were highly expressed on parasitized erythroblasts. As CD8+ T cells recognize MHC class I and peptide complexes on target cells, and are involved in protection or pathology against malaria, we examined whether erythroblasts are targeted by CD8+ T cells. Purified non-parasitized erythroblasts pulsed with OVA peptides were recognized by OVA-specific CD8+ T cells. Crucially, parasitized erythroblasts isolated from GFP-OVA-, but not GFP- infected-mice, activated OT-I CD8+ T cells, indicating that CD8+ T cells recognize parasitized erythroblasts in an antigen-specific manner.
Collapse
Affiliation(s)
- Takashi Imai
- Department of Parasitology, Graduate School of Medicine, Gunma University, Gunma 371-8511, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Hesse C, Ginter W, Förg T, Mayer CT, Baru AM, Arnold-Schrauf C, Unger WWJ, Kalay H, van Kooyk Y, Berod L, Sparwasser T. In vivo targeting of human DC-SIGN drastically enhances CD8⁺ T-cell-mediated protective immunity. Eur J Immunol 2013; 43:2543-53. [PMID: 23784881 DOI: 10.1002/eji.201343429] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Revised: 05/27/2013] [Accepted: 06/17/2013] [Indexed: 11/11/2022]
Abstract
Vaccination is one of the oldest yet still most effective methods to prevent infectious diseases. However, eradication of intracellular pathogens and treatment of certain diseases like cancer requiring efficient cytotoxic immune responses remain a medical challenge. In mice, a successful approach to induce strong cytotoxic CD8⁺ T-cell (CTL) reactions is to target antigens to DCs using specific antibodies against surface receptors in combination with adjuvants. A major drawback for translating this strategy into one for the clinic is the lack of analogous targets in human DCs. DC-SIGN (DC-specific-ICAM3-grabbing-nonintegrin/CD209) is a C-type lectin receptor with potent endocytic capacity and a highly restricted expression on human immature DCs. Therefore, DC-SIGN represents an ideal candidate for DC targeting. Using transgenic mice that express human DC-SIGN under the control of the murine CD11c promoter (hSIGN mice), we explored the efficacy of anti-DC-SIGN antibodies to target antigens to DCs and induce protective immune responses in vivo. We show that anti-DC-SIGN antibodies conjugated to OVA induced strong and persistent antigen-specific CD4⁺ and CD8⁺ T-cell responses, which efficiently protected from infection with OVA-expressing Listeria monocytogenes. Thus, we propose DC targeting via DC-SIGN as a promising strategy for novel vaccination protocols against intracellular pathogens.
Collapse
Affiliation(s)
- Christina Hesse
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Laske K, Shebzukhov YV, Grosse-Hovest L, Kuprash DV, Khlgatian SV, Koroleva EP, Sazykin AY, Penkov DN, Belousov PV, Stevanovic S, Vass V, Walter S, Eisel D, Schmid-Horch BD, Nedospasov SA, Rammensee HG, Gouttefangeas C. Alternative variants of human HYDIN are novel cancer-associated antigens recognized by adaptive immunity. Cancer Immunol Res 2013; 1:190-200. [PMID: 24777681 DOI: 10.1158/2326-6066.cir-13-0079] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A mutation in the hydin gene has been recently described as one possible mechanism leading to lethal congenital hydrocephalus in mice, and a similar defect is proposed to be involved in an autosomal recessive form of hydrocephalus in human. Here, we report for the first time on the cancer association and immunogenicity of two HYDIN variants in humans. One is a previously described sequence derived from the chromosome 1 gene copy, that is, KIAA1864. The second is encoded by a novel alternative transcript originating from the chromosome 16, which we identified by immunoscreening of a testis-derived cDNA expression library with sera of patients with colorectal cancer, and called MO-TES391. Both variants are targeted by immunoglobulin G antibodies in a significant subset of cancer patients but only rarely in healthy donors. Moreover, we identify HLA-A*0201-restricted sequences derived from MO-TES391 and KIAA1864, which are specifically recognized by human cytotoxic CD8(+) T cells. Taken together, these results suggest frequent and coordinated adaptive immune responses against HYDIN variants in patients with cancer and propose HYDIN as a novel cancer-associated antigen.
Collapse
Affiliation(s)
- Karoline Laske
- Authors' Affiliations: Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Ferguson PM, Slocombe A, Tilley RD, Hermans IF. Using magnetic resonance imaging to evaluate dendritic cell-based vaccination. PLoS One 2013; 8:e65318. [PMID: 23734246 PMCID: PMC3667033 DOI: 10.1371/journal.pone.0065318] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Accepted: 04/28/2013] [Indexed: 01/19/2023] Open
Abstract
Cancer immunotherapy with antigen-loaded dendritic cell-based vaccines can induce clinical responses in some patients, but further optimization is required to unlock the full potential of this strategy in the clinic. Optimization is dependent on being able to monitor the cellular events that take place once the dendritic cells have been injected in vivo, and to establish whether antigen-specific immune responses to the tumour have been induced. Here we describe the use of magnetic resonance imaging (MRI) as a simple, non-invasive approach to evaluate vaccine success. By loading the dendritic cells with highly magnetic iron nanoparticles it is possible to assess whether the injected cells drain to the lymph nodes. It is also possible to establish whether an antigen-specific response is initiated by assessing migration of successive rounds of antigen-loaded dendritic cells; in the face of a successfully primed cytotoxic response, the bulk of antigen-loaded cells are eradicated on-route to the node, whereas cells without antigen can reach the node unchecked. It is also possible to verify the induction of a vaccine-induced response by simply monitoring increases in draining lymph node size as a consequence of vaccine-induced lymphocyte trapping, which is an antigen-specific response that becomes more pronounced with repeated vaccination. Overall, these MRI techniques can provide useful early feedback on vaccination strategies, and could also be used in decision making to select responders from non-responders early in therapy.
Collapse
Affiliation(s)
| | - Angela Slocombe
- Department of Radiology, Wellington Hospital, Wellington, New Zealand
| | - Richard D. Tilley
- School of Chemical and Physical Sciences, Victoria University of Wellington, Wellington, New Zealand
| | - Ian F. Hermans
- Malaghan Institute of Medical Research, Wellington, New Zealand
- * E-mail:
| |
Collapse
|
66
|
Strength of PD-1 signaling differentially affects T-cell effector functions. Proc Natl Acad Sci U S A 2013; 110:E2480-9. [PMID: 23610399 DOI: 10.1073/pnas.1305394110] [Citation(s) in RCA: 238] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
High surface expression of programmed death 1 (PD-1) is associated with T-cell exhaustion; however, the relationship between PD-1 expression and T-cell dysfunction has not been delineated. We developed a model to study PD-1 signaling in primary human T cells to study how PD-1 expression affected T-cell function. By determining the number of T-cell receptor/peptide-MHC complexes needed to initiate a Ca(2+) flux, we found that PD-1 ligation dramatically shifts the dose-response curve, making T cells much less sensitive to T-cell receptor-generated signals. Importantly, other T-cell functions were differentially sensitive to PD-1 expression. We observed that high levels of PD-1 expression were required to inhibit macrophage inflammatory protein 1 beta production, lower levels were required to block cytotoxicity and IFN-γ production, and very low levels of PD-1 expression could inhibit TNF-α and IL-2 production as well as T-cell expansion. These findings provide insight into the role of PD-1 expression in enforcing T-cell exhaustion and the therapeutic potential of PD-1 blockade.
Collapse
|
67
|
Hildemann SK, Eberlein J, Davenport B, Nguyen TT, Victorino F, Homann D. High efficiency of antiviral CD4(+) killer T cells. PLoS One 2013; 8:e60420. [PMID: 23565245 PMCID: PMC3614903 DOI: 10.1371/journal.pone.0060420] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 01/10/2013] [Indexed: 12/22/2022] Open
Abstract
The destruction of infected cells by cytotxic T lymphocytes (CTL) is integral to the effective control of viral and bacterial diseases, and CTL function at large has long been regarded as a distinctive property of the CD8(+)T cell subset. In contrast, and despite their first description more than three decades ago, the precise contribution of cytotoxic CD4(+)T cells to the resolution of infectious diseases has remained a matter of debate. In particular, the CTL activity of pathogen-specific CD4(+) "helper" T cells constitutes a single trait among a diverse array of other T cell functionalities, and overall appears considerably weaker than the cytolytic capacity of CD8(+) effector T cells. Here, using an in vivo CTL assay, we report that cytotoxic CD4(+)T cells are readily generated against both viral and bacterial pathogens, and that the efficiency of MHC-II-restricted CD4(+)T cell killing adjusted for effector:target cell ratios, precise specificities and functional avidities is comparable in magnitude to that of CD8(+)T cells. In fact, the only difference between specific CD4(+) and CD8(+)T cells pertains to the slightly delayed killing kinetics of the former demonstrating that potent CTL function is a cardinal property of both antiviral CD8(+) and CD4(+)T cells.
Collapse
Affiliation(s)
- Steven K. Hildemann
- University Clinic for Cardiology and Angiology I, University Heart Center, Freiburg-Bad Krozingen, Germany
- Merck Research Laboratories/MSD Global Clinical Trial Operations, Haar, Germany
| | - Jens Eberlein
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Bennett Davenport
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America
- Integrated Department of Immunology, University of Colorado Denver and National Jewish Health, Denver, Colorado, United States of America
- Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Tom T. Nguyen
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America
- Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Francisco Victorino
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America
- Integrated Department of Immunology, University of Colorado Denver and National Jewish Health, Denver, Colorado, United States of America
| | - Dirk Homann
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America
- Integrated Department of Immunology, University of Colorado Denver and National Jewish Health, Denver, Colorado, United States of America
- Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, United States of America
| |
Collapse
|
68
|
Chu T, Tyznik AJ, Roepke S, Berkley AM, Woodward-Davis A, Pattacini L, Bevan MJ, Zehn D, Prlic M. Bystander-activated memory CD8 T cells control early pathogen load in an innate-like, NKG2D-dependent manner. Cell Rep 2013; 3:701-8. [PMID: 23523350 DOI: 10.1016/j.celrep.2013.02.020] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2012] [Revised: 01/29/2013] [Accepted: 02/19/2013] [Indexed: 02/06/2023] Open
Abstract
During an infection the antigen-nonspecific memory CD8 T cell compartment is not simply an inert pool of cells, but becomes activated and cytotoxic. It is unknown how these cells contribute to the clearance of an infection. We measured the strength of T cell receptor (TCR) signals that bystander-activated, cytotoxic CD8 T cells (BA-CTLs) receive in vivo and found evidence of limited TCR signaling. Given this marginal contribution of the TCR, we asked how BA-CTLs identify infected target cells. We show that target cells express NKG2D ligands following bacterial infection and demonstrate that BA-CTLs directly eliminate these target cells in an innate-like, NKG2D-dependent manner. Selective inhibition of BA-CTL-mediated killing led to a significant defect in pathogen clearance. Together, these data suggest an innate role for memory CD8 T cells in the early immune response before the onset of a de novo generated, antigen-specific CD8 T cell response.
Collapse
Affiliation(s)
- Talyn Chu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
69
|
Quah BJC, Wijesundara DK, Ranasinghe C, Parish CR. Fluorescent target array killing assay: A multiplex cytotoxic T-cell assay to measure detailed T-cell antigen specificity and avidity in vivo. Cytometry A 2012; 81:679-90. [DOI: 10.1002/cyto.a.22084] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 04/20/2012] [Accepted: 05/23/2012] [Indexed: 11/10/2022]
|
70
|
Schmueck M, Fischer AM, Hammoud B, Brestrich G, Fuehrer H, Luu SH, Mueller K, Babel N, Volk HD, Reinke P. Preferential Expansion of Human Virus-Specific Multifunctional Central Memory T Cells by Partial Targeting of the IL-2 Receptor Signaling Pathway: The Key Role of CD4+ T Cells. THE JOURNAL OF IMMUNOLOGY 2012; 188:5189-98. [DOI: 10.4049/jimmunol.1103763] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
71
|
Virus-like particles and α-galactosylceramide form a self-adjuvanting composite particle that elicits anti-tumor responses. J Control Release 2012; 159:338-45. [PMID: 22386518 DOI: 10.1016/j.jconrel.2012.02.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Revised: 02/14/2012] [Accepted: 02/17/2012] [Indexed: 02/08/2023]
Abstract
Virus-like particles (VLP) are effective vehicles for delivery of heterologous antigen to antigen-presenting cells. However VLP alone are insufficiently stimulatory to generate the signals required to facilitate effective priming of naïve T cells. We show that the VLP derived from rabbit hemorrhagic disease virus can bind the galactose-containing adjuvant α-galactosylceramide to form a composite particle for co-delivery of antigen and adjuvant to the same antigen-presenting cell. Vaccination with VLP and α-galactosylceramide activated splenic iNKT cells to produce IFN-γ and IL-4, led to the generation of antigen-specific T cells that protected prophylactically against subcutaneous tumor challenge, and was more effective at generating anti-tumor immune responses than either component individually. These data demonstrate a novel method for immunopotentiating VLP to increase their efficacy in the generation of anti-tumor responses via the innate ligand recognition properties of calicivirus-derived nanoparticles.
Collapse
|
72
|
Flacher V, Tripp CH, Haid B, Kissenpfennig A, Malissen B, Stoitzner P, Idoyaga J, Romani N. Skin langerin+ dendritic cells transport intradermally injected anti-DEC-205 antibodies but are not essential for subsequent cytotoxic CD8+ T cell responses. THE JOURNAL OF IMMUNOLOGY 2012; 188:2146-55. [PMID: 22291181 DOI: 10.4049/jimmunol.1004120] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Incorporation of Ags by dendritic cells (DCs) increases when Ags are targeted to endocytic receptors by mAbs. We have previously demonstrated in the mouse that mAbs against C-type lectins administered intradermally are taken up by epidermal Langerhans cells (LCs), dermal Langerin(neg) DCs, and dermal Langerin(+) DCs in situ. However, the relative contribution of these skin DC subsets to the induction of immune responses after Ag targeting has not been addressed in vivo. We show in this study that murine epidermal LCs and dermal DCs transport intradermally injected mAbs against the lectin receptor DEC-205/CD205 in vivo. Skin DCs targeted in situ with mAbs migrated through lymphatic vessels in steady state and inflammation. In the skin-draining lymph nodes, targeting mAbs were found in resident CD8α(+) DCs and in migrating skin DCs. More than 70% of targeted DCs expressed Langerin, including dermal Langerin(+) DCs and LCs. Numbers of targeted skin DCs in the nodes increased 2-3-fold when skin was topically inflamed by the TLR7 agonist imiquimod. Complete removal of the site where OVA-coupled anti-DEC-205 had been injected decreased endogenous cytotoxic responses against OVA peptide-loaded target cells by 40-50%. Surprisingly, selective ablation of all Langerin(+) skin DCs in Langerin-DTR knock-in mice did not affect such responses independently of the adjuvant chosen. Thus, in cutaneous immunization strategies where Ag is targeted to DCs, Langerin(+) skin DCs play a major role in transport of anti-DEC-205 mAb, although Langerin(neg) dermal DCs and CD8α(+) DCs are sufficient to subsequent CD8(+) T cell responses.
Collapse
Affiliation(s)
- Vincent Flacher
- Department of Dermatology and Venereology, Innsbruck Medical University, A-6020 Innsbruck, Austria
| | | | | | | | | | | | | | | |
Collapse
|
73
|
Tissue exit: a novel control point in the accumulation of antigen-specific CD8 T cells in the influenza a virus-infected lung. J Virol 2012; 86:3436-45. [PMID: 22278253 DOI: 10.1128/jvi.07025-11] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Memory/effector T cells efficiently migrate into extralymphoid tissues and sites of infection, providing immunosurveillance and a first line of defense against invading pathogens. Even though it is a potential means to regulate the size, quality, and duration of a tissue infiltrate, T cell egress from infected tissues is poorly understood. Using a mouse model of influenza A virus infection, we found that CD8 effector T cells egressed from the infected lung in a CCR7-dependent manner. In contrast, following antigen recognition, effector CD8 T cell egress decreased and CCR7 function was reduced in vivo and in vitro, indicating that the exit of CD8 T cells from infected tissues is tightly regulated. Our data suggest that the regulation of T cell egress is a mechanism to retain antigen-specific effectors at the site of infection to promote viral clearance, while decreasing the numbers of bystander T cells and preventing overt inflammation.
Collapse
|
74
|
Barrett DM, Zhao Y, Liu X, Jiang S, Carpenito C, Kalos M, Carroll RG, June CH, Grupp SA. Treatment of advanced leukemia in mice with mRNA engineered T cells. Hum Gene Ther 2011; 22:1575-86. [PMID: 21838572 DOI: 10.1089/hum.2011.070] [Citation(s) in RCA: 175] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cytotoxic T lymphocytes (CTLs) modified with chimeric antigen receptors (CARs) for adoptive immunotherapy of hematologic malignancies are effective in preclinical models and are being tested in several clinical trials. Although CTLs bearing stably expressed CARs generated by integrating viral vectors are efficacious and have potential long-term persistence, this mechanism of CAR expression can potentially result in significant toxicity. T cells were electroporated with an optimized in vitro transcribed RNA encoding a CAR against CD19. These RNA CAR CTLs were then tested in vitro and in vivo for efficacy. We found that T cells expressing an anti-CD19 CAR introduced by electroporation with optimized mRNA were potent and specific killers of CD19 target cells. CD19 RNA CAR T cells given to immunodeficient mice bearing xenografted leukemia rapidly migrated to sites of disease and retained significant target-specific lytic activity. Unexpectedly, a single injection of CD19 RNA CAR T cells reduced disease burden within 1 day after administration, resulting in a significant prolongation of survival in an aggressive leukemia xenograft model. The surface expression of the RNA CARs may be titrated, giving T cells with potentially tunable levels of effector functions such as cytokine release and cytotoxicity. RNA CARs are a genetic engineering approach that should not be subject to genotoxicity, and they provide a platform for rapidly optimizing CAR design before proceeding to more costly and laborious stable expression systems.
Collapse
Affiliation(s)
- David M Barrett
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Pre-clinical efficacy and safety of experimental vaccines based on non-replicating vaccinia vectors against yellow fever. PLoS One 2011; 6:e24505. [PMID: 21931732 PMCID: PMC3170363 DOI: 10.1371/journal.pone.0024505] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Accepted: 08/12/2011] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Currently existing yellow fever (YF) vaccines are based on the live attenuated yellow fever virus 17D strain (YFV-17D). Although, a good safety profile was historically attributed to the 17D vaccine, serious adverse events have been reported, making the development of a safer, more modern vaccine desirable. METHODOLOGY/PRINCIPAL FINDINGS A gene encoding the precursor of the membrane and envelope (prME) protein of the YFV-17D strain was inserted into the non-replicating modified vaccinia virus Ankara and into the D4R-defective vaccinia virus. Candidate vaccines based on the recombinant vaccinia viruses were assessed for immunogenicity and protection in a mouse model and compared to the commercial YFV-17D vaccine. The recombinant live vaccines induced γ-interferon-secreting CD4- and functionally active CD8-T cells, and conferred full protection against lethal challenge already after a single low immunization dose of 10(5) TCID(50). Surprisingly, pre-existing immunity against wild-type vaccinia virus did not negatively influence protection. Unlike the classical 17D vaccine, the vaccinia virus-based vaccines did not cause mortality following intracerebral administration in mice, demonstrating better safety profiles. CONCLUSIONS/SIGNIFICANCE The non-replicating recombinant YF candidate live vaccines induced a broad immune response after single dose administration, were effective even in the presence of a pre-existing immunity against vaccinia virus and demonstrated an excellent safety profile in mice.
Collapse
|
76
|
Crawford A, Angelosanto JM, Nadwodny KL, Blackburn SD, Wherry EJ. A role for the chemokine RANTES in regulating CD8 T cell responses during chronic viral infection. PLoS Pathog 2011; 7:e1002098. [PMID: 21814510 PMCID: PMC3141034 DOI: 10.1371/journal.ppat.1002098] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Accepted: 04/15/2011] [Indexed: 01/22/2023] Open
Abstract
RANTES (CCL5) is a chemokine expressed by many hematopoietic and non-hematopoietic cell types that plays an important role in homing and migration of effector and memory T cells during acute infections. The RANTES receptor, CCR5, is a major target of anti-HIV drugs based on blocking viral entry. However, defects in RANTES or RANTES receptors including CCR5 can compromise immunity to acute infections in animal models and lead to more severe disease in humans infected with west Nile virus (WNV). In contrast, the role of the RANTES pathway in regulating T cell responses and immunity during chronic infection remains unclear. In this study, we demonstrate a crucial role for RANTES in the control of systemic chronic LCMV infection. In RANTES−/− mice, virus-specific CD8 T cells had poor cytokine production. These RANTES−/− CD8 T cells also expressed higher amounts of inhibitory receptors consistent with more severe exhaustion. Moreover, the cytotoxic ability of CD8 T cells from RANTES−/− mice was reduced. Consequently, viral load was higher in the absence of RANTES. The dysfunction of T cells in the absence of RANTES was as severe as CD8 T cell responses generated in the absence of CD4 T cell help. Our results demonstrate an important role for RANTES in sustaining CD8 T cell responses during a systemic chronic viral infection. Chemokines are small proteins that attract cells and play complex roles in coordinating immune responses. RANTES is one such chemokine that attracts many different cell types. The receptor for RANTES, CCR5, is also a coreceptor for HIV and drugs blocking the RANTES∶CCR5 pathway are in clinical use to treat HIV-infected individuals. Despite the importance of CCR5 during HIV infection, the role of RANTES during other chronic infections remains poorly defined. In this study, we found that the absence of RANTES limited the ability of mice to control chronic LCMV infection resulting in higher viral loads and more severe T cell exhaustion. Our data suggest that the impact of blocking the RANTES∶CCR5 pathway on the ability to control other chronic infections should be given careful consideration when treating HIV-infected individuals.
Collapse
Affiliation(s)
- Alison Crawford
- Department of Microbiology and Institute for Immunology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Jill Marie Angelosanto
- Department of Microbiology and Institute for Immunology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Kim Lynn Nadwodny
- GlaxoSmithKline, Department of Safety Assessment, Immunologic Toxicology, King of Prussia, Pennsylvania, United States of America
| | - Shawn D. Blackburn
- Department of Microbiology and Institute for Immunology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - E. John Wherry
- Department of Microbiology and Institute for Immunology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
77
|
Duewell P, Kisser U, Heckelsmiller K, Hoves S, Stoitzner P, Koernig S, Morelli AB, Clausen BE, Dauer M, Eigler A, Anz D, Bourquin C, Maraskovsky E, Endres S, Schnurr M. ISCOMATRIX adjuvant combines immune activation with antigen delivery to dendritic cells in vivo leading to effective cross-priming of CD8+ T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 187:55-63. [PMID: 21613613 PMCID: PMC4285562 DOI: 10.4049/jimmunol.1004114] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cancer vaccines aim to induce CTL responses against tumors. Challenges for vaccine design are targeting Ag to dendritic cells (DCs) in vivo, facilitating cross-presentation, and conditioning the microenvironment for Th1 type immune responses. In this study, we report that ISCOM vaccines, which consist of ISCOMATRIX adjuvant and protein Ag, meet these challenges. Subcutaneous injection of an ISCOM vaccine in mice led to a substantial influx and activation of innate and adaptive immune effector cells in vaccine site-draining lymph nodes (VDLNs) as well as IFN-γ production by NK and NKT cells. Moreover, an ISCOM vaccine containing the model Ag OVA (OVA/ISCOM vaccine) was efficiently taken up by CD8α(+) DCs in VDLNs and induced their maturation and IL-12 production. Adoptive transfer of transgenic OT-I T cells revealed highly efficient cross-presentation of the OVA/ISCOM vaccine in vivo, whereas cross-presentation of soluble OVA was poor even at a 100-fold higher concentration. Cross-presenting activity was restricted to CD8α(+) DCs in VDLNs, whereas Langerin(+) DCs and CD8α(-) DCs were dispensable. Remarkably, compared with other adjuvant systems, the OVA/ISCOM vaccine induced a high frequency of OVA-specific CTLs capable of tumor cell killing in different tumor models. Thus, ISCOM vaccines combine potent immune activation with Ag delivery to CD8α(+) DCs in vivo for efficient induction of CTL responses.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cell Line, Tumor
- Cells, Cultured
- Cholesterol/administration & dosage
- Cholesterol/immunology
- Cross-Priming/immunology
- Cytotoxicity Tests, Immunologic/methods
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Drug Combinations
- Female
- Gene Knock-In Techniques
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Killer Cells, Natural/pathology
- Lymph Nodes/immunology
- Lymph Nodes/pathology
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/prevention & control
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Natural Killer T-Cells/immunology
- Natural Killer T-Cells/metabolism
- Natural Killer T-Cells/pathology
- Ovalbumin/administration & dosage
- Ovalbumin/immunology
- Peptide Fragments/administration & dosage
- Peptide Fragments/immunology
- Phospholipids/administration & dosage
- Phospholipids/immunology
- Quillaja/immunology
- Saponins/administration & dosage
- Saponins/immunology
Collapse
Affiliation(s)
- Peter Duewell
- Medizinische Klinik Innenstadt, Ludwig-Maximilians-Universität, 80336 Munich, Germany
| | - Ulrich Kisser
- Medizinische Klinik Innenstadt, Ludwig-Maximilians-Universität, 80336 Munich, Germany
| | - Klaus Heckelsmiller
- Medizinische Klinik Innenstadt, Ludwig-Maximilians-Universität, 80336 Munich, Germany
| | - Sabine Hoves
- Medizinische Klinik Innenstadt, Ludwig-Maximilians-Universität, 80336 Munich, Germany
| | - Patrizia Stoitzner
- Department of Dermatology, Innsbruck Medical University, 6020 Innsbruck, Austria
| | | | | | - Björn E. Clausen
- Department of Immunology, Erasmus Medical Center, University Medical Center, 3015 GE Rotterdam, The Netherlands
| | - Marc Dauer
- Department of Medicine II, Saarland University Hospital, 66421 Homburg, Germany
| | - Andreas Eigler
- Klinik für Innere Medizin I, Klinikum Dritter Orden, 80638 Munich, Germany
| | - David Anz
- Division of Clinical Pharmacology, Ludwig-Maximilians University, 80336 Munich, Germany
| | - Carole Bourquin
- Division of Clinical Pharmacology, Ludwig-Maximilians University, 80336 Munich, Germany
| | | | - Stefan Endres
- Division of Clinical Pharmacology, Ludwig-Maximilians University, 80336 Munich, Germany
| | - Max Schnurr
- Medizinische Klinik Innenstadt, Ludwig-Maximilians-Universität, 80336 Munich, Germany
| |
Collapse
|
78
|
Ebensen T, Libanova R, Schulze K, Yevsa T, Morr M, Guzmán CA. Bis-(3',5')-cyclic dimeric adenosine monophosphate: strong Th1/Th2/Th17 promoting mucosal adjuvant. Vaccine 2011; 29:5210-20. [PMID: 21619907 DOI: 10.1016/j.vaccine.2011.05.026] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Revised: 04/07/2011] [Accepted: 05/11/2011] [Indexed: 12/24/2022]
Abstract
New effective adjuvants are required to improve the performance of subunit vaccines. Here, we showed that bis-(3',5')-cyclic dimeric adenosine monophosphate (c-di-AMP), a second messenger molecule in bacteria and archaea, exerts strong adjuvant activities when delivered by mucosal route. In vitro studies showed that c-di-AMP was able to both stimulate pre-activated murine macrophages and promote the activation and maturation of dendritic cells of murine and human origin. Co-administration of c-di-AMP with β-galactosidase (β-Gal) by intranasal route to BALB/c mice resulted in the elicitation of significantly higher serum antigen-specific IgG titres than in controls. The induction of local immune responses was shown by the production of antigen-specific secretory IgA in different mucosal territories. In addition, strong cellular immune responses were observed against both the β-Gal protein and a peptide encompassing its MHC class I-restricted epitope. The ratio of β-Gal-specific antibodies and the secreted cytokine profiles by in vitro re-stimulated splenocytes suggested that a balanced Th1/Th2/Th17 response pattern is promoted by c-di-AMP. When C57BL/6 mice were immunized with OVA and c-di-AMP, vigorous in vivo CTL responses were also observed. These results indicated that c-di-AMP exhibits a high potential as adjuvant for the development of mucosal vaccines, in particular when cellular immunity is needed.
Collapse
Affiliation(s)
- Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany.
| | | | | | | | | | | |
Collapse
|
79
|
Hopkins R, Bridgeman A, Joseph J, Gilbert SC, McShane H, Hanke T. Dual neonate vaccine platform against HIV-1 and M. tuberculosis. PLoS One 2011; 6:e20067. [PMID: 21603645 PMCID: PMC3094449 DOI: 10.1371/journal.pone.0020067] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Accepted: 04/20/2011] [Indexed: 01/05/2023] Open
Abstract
Acquired immunodeficiency syndrome and tuberculosis (TB) are two of the
world's most devastating diseases. The first vaccine the majority of
infants born in Africa receive is Mycobacterium bovis bacillus
Calmette-Guérin (BCG) as a prevention against TB. BCG protects against
disseminated disease in the first 10 years of life, but provides a variable
protection against pulmonary TB and enhancing boost delivered by recombinant
modified vaccinia virus Ankara (rMVA) expressing antigen 85A (Ag85A) of
M. tuberculosis is currently in phase IIb evaluation in
African neonates. If the newborn's mother is positive for human
immunodeficiency virus type 1 (HIV-1), the baby is at high risk of acquiring
HIV-1 through breastfeeding. We suggested that a vaccination consisting of
recombinant BCG expressing HIV-1 immunogen administered at birth followed by a
boost with rMVA sharing the same immunogen could serve as a strategy for
prevention of mother-to-child transmission of HIV-1 and rMVA expressing an
African HIV-1-derived immunogen HIVA is currently in phase I trials in African
neonates. Here, we aim to develop a dual neonate vaccine platform against HIV-1
and TB consisting of BCG.HIVA administered at birth followed by a boost with
MVA.HIVA.85A. Thus, mMVA.HIVA.85A and sMVA.HIVA.85A vaccines were constructed,
in which the transgene transcription is driven by either modified H5 or short
synthetic promoters, respectively, and tested for immunogenicity alone and in
combination with BCG.HIVA222. mMVA.HIVA.85A was produced markerless
and thus suitable for clinical manufacture. While sMVA.HIVA.85A expressed higher
levels of the immunogens, it was less immunogenic than mMVA.HIVA.85A in BALB/c
mice. A BCG.HIVA222–mMVA.HIVA.85A prime-boost regimen induced
robust T cell responses to both HIV-1 and M. tuberculosis.
Therefore, proof-of-principle for a dual anti-HIV-1/M.
tuberculosis infant vaccine platform is established. Induction of
immune responses against these pathogens soon after birth is highly desirable
and may provide a basis for lifetime protection maintained by boosts later in
life.
Collapse
Affiliation(s)
- Richard Hopkins
- MRC Human Immunology Unit, Weatherall
Institute of Molecular Medicine, University of Oxford, Oxford, United
Kingdom
| | - Anne Bridgeman
- MRC Human Immunology Unit, Weatherall
Institute of Molecular Medicine, University of Oxford, Oxford, United
Kingdom
| | - Joan Joseph
- AIDS Research Unit, Hospital
Clínic/IDIBAPS-HIVACAT, School of Medicine, University of Barcelona,
Barcelona, Spain
| | - Sarah C. Gilbert
- The Jenner Institute, University of Oxford,
Oxford, United Kingdom
| | - Helen McShane
- The Jenner Institute, University of Oxford,
Oxford, United Kingdom
| | - Tomáš Hanke
- MRC Human Immunology Unit, Weatherall
Institute of Molecular Medicine, University of Oxford, Oxford, United
Kingdom
- The Jenner Institute, University of Oxford,
Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
80
|
Im EJ, Hong JP, Roshorm Y, Bridgeman A, Létourneau S, Liljeström P, Potash MJ, Volsky DJ, McMichael AJ, Hanke T. Protective efficacy of serially up-ranked subdominant CD8+ T cell epitopes against virus challenges. PLoS Pathog 2011; 7:e1002041. [PMID: 21625575 PMCID: PMC3098219 DOI: 10.1371/journal.ppat.1002041] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 03/08/2011] [Indexed: 12/20/2022] Open
Abstract
Immunodominance in T cell responses to complex antigens like viruses is still incompletely understood. Some data indicate that the dominant responses to viruses are not necessarily the most protective, while other data imply that dominant responses are the most important. The issue is of considerable importance to the rational design of vaccines, particularly against variable escaping viruses like human immunodeficiency virus type 1 and hepatitis C virus. Here, we showed that sequential inactivation of dominant epitopes up-ranks the remaining subdominant determinants. Importantly, we demonstrated that subdominant epitopes can induce robust responses and protect against whole viruses if they are allowed at least once in the vaccination regimen to locally or temporally dominate T cell induction. Therefore, refocusing T cell immune responses away from highly variable determinants recognized during natural virus infection towards subdominant, but conserved regions is possible and merits evaluation in humans.
Collapse
Affiliation(s)
- Eung-Jun Im
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe Hospital, Oxford, United Kingdom
| | - Jessie P. Hong
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe Hospital, Oxford, United Kingdom
| | - Yaowaluck Roshorm
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe Hospital, Oxford, United Kingdom
| | - Anne Bridgeman
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe Hospital, Oxford, United Kingdom
| | - Sven Létourneau
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe Hospital, Oxford, United Kingdom
| | - Peter Liljeström
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Mary Jane Potash
- Molecular Virology Division, St. Luke's Roosevelt Hospital Center, Columbia University Medical Center, New York, New York, United States of America
| | - David J. Volsky
- Molecular Virology Division, St. Luke's Roosevelt Hospital Center, Columbia University Medical Center, New York, New York, United States of America
| | - Andrew J. McMichael
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe Hospital, Oxford, United Kingdom
| | - Tomáš Hanke
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, The John Radcliffe Hospital, Oxford, United Kingdom
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
81
|
Saubi N, Im EJ, Fernández-Lloris R, Gil O, Cardona PJ, Gatell JM, Hanke T, Joseph J. Newborn mice vaccination with BCG.HIVA²²² + MVA.HIVA enhances HIV-1-specific immune responses: influence of age and immunization routes. Clin Dev Immunol 2011; 2011:516219. [PMID: 21603216 PMCID: PMC3095426 DOI: 10.1155/2011/516219] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Accepted: 02/01/2011] [Indexed: 11/17/2022]
Abstract
We have evaluated the influence of age and immunization routes for induction of HIV-1- and M. tuberculosis-specific immune responses after neonatal (7 days old) and adult (7 weeks old) BALB/c mice immunization with BCG.HIVA(222) prime and MVA.HIVA boost. The specific HIV-1 cellular immune responses were analyzed in spleen cells. The body weight of the newborn mice was weekly recorded. The frequencies of HIV-specific CD8(+) T cells producing IFN-γ were higher in adult mice vaccinated intradermally and lower in adult and newborn mice vaccinated subcutaneously. In all cases the IFN-γ production was significantly higher when mice were primed with BCG.HIVA(222) compared with BCGwt. When the HIV-specific CTL activity was assessed, the frequencies of specific killing were higher in newborn mice than in adults. The prime-boost vaccination regimen which includes BCG.HIVA(222) and MVA.HIVA was safe when inoculated to newborn mice. The administration of BCG.HIVA(222) to newborn mice is safe and immunogenic and increased the HIV-specific responses induced by MVA.HIVA vaccine. It might be a good model for infant HIV and Tuberculosis bivalent vaccine.
Collapse
Affiliation(s)
- Narcís Saubi
- AIDS Research Unit, Hospital Clínic/IDIBAPS-HIVACAT, University of Barcelona, Calle Villarroel 170, 08036 Barcelona, Spain
| | - Eung-Jun Im
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Oxford University and The John Radcliffe, Oxford OX3 9DS, UK
| | - Raquel Fernández-Lloris
- AIDS Research Unit, Hospital Clínic/IDIBAPS-HIVACAT, University of Barcelona, Calle Villarroel 170, 08036 Barcelona, Spain
| | - Olga Gil
- Unitat Tuberculosi Experimental, Institut “Germans Trias i Pujol”, Carretera del Canyet S/N, Badalona 08916, Barcelona, Spain
| | - Pere-Joan Cardona
- Unitat Tuberculosi Experimental, Institut “Germans Trias i Pujol”, Carretera del Canyet S/N, Badalona 08916, Barcelona, Spain
| | - Josep Maria Gatell
- AIDS Research Unit, Hospital Clínic/IDIBAPS-HIVACAT, University of Barcelona, Calle Villarroel 170, 08036 Barcelona, Spain
| | - Tomáš Hanke
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Oxford University and The John Radcliffe, Oxford OX3 9DS, UK
| | - Joan Joseph
- AIDS Research Unit, Hospital Clínic/IDIBAPS-HIVACAT, University of Barcelona, Calle Villarroel 170, 08036 Barcelona, Spain
| |
Collapse
|
82
|
Lutz‐Nicoladoni C, Wallner S, Stoitzner P, Pircher M, Gruber T, Wolf AM, Gastl G, Penninger JM, Baier G, Wolf D. Reinforcement of cancer immunotherapy by adoptive transfer of
cblb
‐deficient CD8
+
T cells combined with a DC vaccine. Immunol Cell Biol 2011; 90:130-4. [DOI: 10.1038/icb.2011.11] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Christina Lutz‐Nicoladoni
- Tyrolean Cancer Research Institute, Innsbruck Medical University Tyrol Austria
- Internal Medicine V, Hematology and Oncology, Innsbruck Medical University Tyrol Austria
| | - Stephanie Wallner
- Tyrolean Cancer Research Institute, Innsbruck Medical University Tyrol Austria
- Internal Medicine V, Hematology and Oncology, Innsbruck Medical University Tyrol Austria
| | - Patrizia Stoitzner
- Department of Dermatology and Venereology, Innsbruck Medical University Tyrol Austria
| | - Magdalena Pircher
- Tyrolean Cancer Research Institute, Innsbruck Medical University Tyrol Austria
- Internal Medicine V, Hematology and Oncology, Innsbruck Medical University Tyrol Austria
| | - Thomas Gruber
- Department for Medical Genetics, Molecular and Clinical Pharmacology, Innsbruck Medical University Tyrol Austria
| | - Anna Maria Wolf
- Tyrolean Cancer Research Institute, Innsbruck Medical University Tyrol Austria
- Internal Medicine V, Hematology and Oncology, Innsbruck Medical University Tyrol Austria
| | - Günther Gastl
- Tyrolean Cancer Research Institute, Innsbruck Medical University Tyrol Austria
- Internal Medicine V, Hematology and Oncology, Innsbruck Medical University Tyrol Austria
| | - Josef M Penninger
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences Vienna Austria
| | - Gottfried Baier
- Department for Medical Genetics, Molecular and Clinical Pharmacology, Innsbruck Medical University Tyrol Austria
| | - Dominik Wolf
- Tyrolean Cancer Research Institute, Innsbruck Medical University Tyrol Austria
- Internal Medicine V, Hematology and Oncology, Innsbruck Medical University Tyrol Austria
| |
Collapse
|
83
|
Ganusov VV, Barber DL, De Boer RJ. Killing of targets by CD8 T cells in the mouse spleen follows the law of mass action. PLoS One 2011; 6:e15959. [PMID: 21283669 PMCID: PMC3025913 DOI: 10.1371/journal.pone.0015959] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Accepted: 12/01/2010] [Indexed: 12/11/2022] Open
Abstract
It has been difficult to correlate the quality of CD8 T cell responses with protection against viral infections. To investigate the relationship between efficacy and magnitude of T cell responses, we quantify the rate at which individual CD8 effector and memory T cells kill target cells in the mouse spleen. Using mathematical modeling, we analyze recent data on the loss of target cells pulsed with three different peptides from the mouse lymphocytic choriomeningitis virus (LCMV) in mouse spleens with varying numbers of epitope-specific CD8 T cells. We find that the killing of targets follows the law of mass-action, i.e., the death rate of individual target cells remains proportional to the frequency (or the total number) of specific CD8 T cells in the spleen despite the fact that effector cell densities and effector to target ratios vary about a 1000-fold. The killing rate of LCMV-specific CD8 T cells is largely independent of T cell specificity and differentiation stage. Our results thus allow one to calculate the critical T cell concentration at which growth of a virus with a given replication rate can be prevented from the start of infection by memory CD8 T cell response.
Collapse
Affiliation(s)
- Vitaly V. Ganusov
- Department of Microbiology, University of Tennessee, Knoxville, Tennessee, United States of America
- * E-mail:
| | - Daniel L. Barber
- National Institutes of Health, Bethesda, Massachusetts, United States of America
| | - Rob J. De Boer
- Theoretical Biology, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
84
|
Mikkelsen M, Holst PJ, Bukh J, Thomsen AR, Christensen JP. Enhanced and Sustained CD8+T Cell Responses with an Adenoviral Vector-Based Hepatitis C Virus Vaccine Encoding NS3 Linked to the MHC Class II Chaperone Protein Invariant Chain. THE JOURNAL OF IMMUNOLOGY 2011; 186:2355-64. [DOI: 10.4049/jimmunol.1001877] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
85
|
McMahan RH, Golden-Mason L, Nishimura MI, McMahon BJ, Kemper M, Allen TM, Gretch DR, Rosen HR. Tim-3 expression on PD-1+ HCV-specific human CTLs is associated with viral persistence, and its blockade restores hepatocyte-directed in vitro cytotoxicity. J Clin Invest 2010; 120:4546-57. [PMID: 21084749 DOI: 10.1172/jci43127] [Citation(s) in RCA: 259] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Accepted: 09/22/2010] [Indexed: 12/16/2022] Open
Abstract
Having successfully developed mechanisms to evade immune clearance, hepatitis C virus (HCV) establishes persistent infection in approximately 75%-80% of patients. In these individuals, the function of HCV-specific CD8+ T cells is impaired by ligation of inhibitory receptors, the repertoire of which has expanded considerably in the past few years. We hypothesized that the coexpression of the negative regulatory receptors T cell immunoglobulin and mucin domain-containing molecule 3 (Tim-3) and programmed death 1 (PD-1) in HCV infection would identify patients at risk of developing viral persistence during and after acute HCV infection. The frequency of PD-1-Tim-3- HCV-specific CTLs greatly outnumbered PD-1+Tim-3+ CTLs in patients with acute resolving infection. Moreover, the population of PD-1+Tim-3+ T cells was enriched for within the central memory T cell subset and within the liver. Blockade of either PD-1 or Tim-3 enhanced in vitro proliferation of HCV-specific CTLs to a similar extent, whereas cytotoxicity against a hepatocyte cell line that expressed cognate HCV epitopes was increased exclusively by Tim-3 blockade. These results indicate that the coexpression of these inhibitory molecules tracks with defective T cell responses and that anatomical differences might account for lack of immune control of persistent pathogens, which suggests their manipulation may represent a rational target for novel immunotherapeutic approaches.
Collapse
Affiliation(s)
- Rachel H McMahan
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Colorado Denver, Denver, Colorado, USA
| | | | | | | | | | | | | | | |
Collapse
|
86
|
Cao LF, Krymskaya L, Tran V, Mi S, Jensen MC, Blanchard S, Kalos M. Development and application of a multiplexable flow cytometry-based assay to quantify cell-mediated cytolysis. Cytometry A 2010; 77:534-45. [PMID: 20229499 DOI: 10.1002/cyto.a.20887] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Although target cell cytolysis has been widely employed to describe effector function of cells, cytolysis assays as commonly employed do not generate quantitative data. In this report we describe the development and application of a statistically supported flow cytometry-based assay to quantify cell-mediated cytolysis. The assay depends on the use of the fluorescent dye CFSE to distinguish target from effector cells, the DNA intercalating dye 7AAD to distinguish dead from live cell events, and on the establishment of a cytolysis curve that allows for the derivation of statistically robust data. We demonstrate that the cytolysis curve is well described by a four parameter logistic regression model provided that (i) the range of effector to target (E:T) ratios studied allows for full description of the logistic curve, and (ii) an adequate number of data points are collected to estimate the model parameters. We show that the assay is highly reproducible and accurate, and comparable in sensitivity with the standard (51)Cr assay. We report on the potential for this assay to generate quantitative data on the cytolytic activity of both CD8 T and NK cells; describe a relationship between the efficiency of effector cell degranulation and target cell cytolysis throughout a range of E:T ratios, and demonstrate the potential to multiplex with other platforms to obtain broader datasets for the effector phenotype of cells. Appropriate use of this assay will enhance the ability to derive quantitative and integrated correlative datasets from basic, translational, and clinical studies.
Collapse
Affiliation(s)
- Lan-Feng Cao
- Clinical Immunobiology Correlative Studies Laboratory, City of Hope Duarte, California, USA
| | | | | | | | | | | | | |
Collapse
|
87
|
Wingender G, Krebs P, Beutler B, Kronenberg M. Antigen-specific cytotoxicity by invariant NKT cells in vivo is CD95/CD178-dependent and is correlated with antigenic potency. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 185:2721-9. [PMID: 20660713 PMCID: PMC2989418 DOI: 10.4049/jimmunol.1001018] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Invariant NKT (iNKT) cells are a unique subset of T lymphocytes that rapidly carry out effector functions following activation with glycolipid Ags, such as the model Ag alpha-galactosylceramide. Numerous studies have investigated the mechanisms leading to Th1 and Th2 cytokine production by iNKT cells, as well as the effects of the copious amounts of cytokines these cells produce. Less is known, however, about the mechanisms of iNKT cell cytotoxicity. In this study, we investigated the effect of Ag availability and strength, as well as the molecules involved in iNKT cytotoxicity. We demonstrate that the iNKT cell cytotoxicity in vivo correlates directly with the amount of CD1d expressed by the targets as well as the TCR affinity for the target glycolipid Ag. iNKT cells from spleen, liver, and thymus were comparable in their cytotoxicity in vitro. Surprisingly, we show that the Ag-specific cytotoxicity of iNKT cells in vivo depended almost exclusively on the interaction of CD95 (Fas) with CD178 (FasL), and that this mechanism can be efficiently used for tumor protection. Therefore, unlike NK cells, which rely mostly on perforin/granzyme-mediated mechanisms, the Ag-specific cytotoxicity of iNKT cells in vivo is largely restricted to the CD95/CD178 pathway.
Collapse
MESH Headings
- Animals
- Antigens, CD1d/biosynthesis
- Antigens, CD1d/physiology
- Cell Line, Tumor
- Cytotoxicity Tests, Immunologic/methods
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/toxicity
- Fas Ligand Protein/metabolism
- Fas Ligand Protein/physiology
- Galactosylceramides/toxicity
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Lung Neoplasms/therapy
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Natural Killer T-Cells/immunology
- Natural Killer T-Cells/metabolism
- Natural Killer T-Cells/pathology
- Up-Regulation/immunology
- fas Receptor/metabolism
- fas Receptor/physiology
Collapse
Affiliation(s)
- Gerhard Wingender
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Philippe Krebs
- Department of Genetics, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Bruce Beutler
- Department of Genetics, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Mitchell Kronenberg
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| |
Collapse
|
88
|
Stanke J, Hoffmann C, Erben U, von Keyserling H, Stevanovic S, Cichon G, Schneider A, Kaufmann AM. A flow cytometry-based assay to assess minute frequencies of CD8+ T cells by their cytolytic function. J Immunol Methods 2010; 360:56-65. [PMID: 20558172 DOI: 10.1016/j.jim.2010.06.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Revised: 05/20/2010] [Accepted: 06/07/2010] [Indexed: 10/19/2022]
Abstract
Limited sample size and low sensitivity of currently used functional assays challenge direct analysis of cytotoxic CD8+ T lymphocyte activity to quantify antigen-specific immunity after infection or vaccination. Our flow cytometry-based assay reproducibly detects at least three epitope-specific CD8+ T lymphocytes by their cytolytic function. As exemplified for viral epitopes restricted to the human leukocyte antigen (HLA)-A2, the HLA-A2+ human somatic cell hybrid T2 provided an about 10-fold more sensitive readout as compared to autologous B-lymphoblastoid cells or the human erythroleukemia cell line K562 transfected to express HLA-A2 when used as target cells. We named our assay VITAL-FR assay, referring to Hermans et al. (2004) and indicating the modification of using Far Red (FR) dye instead of CMTMR. Under optimal conditions the VITAL-FR assay proved 30 times more sensitive than the 51chromium-release assay to assess epitope-specific target cell lysis. The high overall sensitivity of the VITAL-FR assay basically depended on the negligible spectral overlap of the emission of a stable Far Red fluorescent reporter with the green tracer for target cell labelling. It also profited from long co-incubation of effector and target cells of up to 72, from prior in-vitro culture increasing the frequency of epitope-specific CD8+ T cells and from generic, easily accessible standardized target cells that were used with only 10(3) specific and 10(3) control target cells per individual experimental reaction. Our functional approach with the VITAL-FR assay therefore ideally suits for monitoring CD8+ T cell-mediated cytotoxicity in e.g. vaccination studies with known MHC-restricted immunogenic peptides in scientific and diagnostic applications.
Collapse
Affiliation(s)
- Jonas Stanke
- Gynecology, Gynecologic Tumor Immunology, Campus Benjamin Franklin and Mitte, Charité-Universitätsmedizin, Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
89
|
Zaritskaya L, Shurin MR, Sayers TJ, Malyguine AM. New flow cytometric assays for monitoring cell-mediated cytotoxicity. Expert Rev Vaccines 2010; 9:601-16. [PMID: 20518716 PMCID: PMC2911950 DOI: 10.1586/erv.10.49] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The exact immunologic responses after vaccination that result in effective antitumor immunity have not yet been fully elucidated and the data from ex vivo T-cell assays have not yet defined adequate surrogate markers for clinical efficacy. A more detailed knowledge of the specific immune responses that correlate with positive clinical outcomes should help to develop better or novel strategies to effectively activate the immune system against tumors. Furthermore, clinically relevant material is often limited and, thus, precludes the ability to perform multiple assays. The two main assays currently used to monitor lymphocyte-mediated cytoxicity in cancer patients are the (51)Cr-release assay and IFN-gamma ELISpot assay. The former has a number of disadvantages, including low sensitivity, poor labeling and high spontaneous release of isotope from some tumor target cells. Additional problems with the (51)Cr-release assay include difficulty in obtaining autologous tumor targets, and biohazard and disposal problems for the isotope. The ELISpot assays do not directly measure cytotoxic activity and are, therefore, a surrogate marker of cyotoxic capacity of effector T cells. Furthermore, they do not assess cytotoxicity mediated by the production of the TNF family of death ligands by the cytotoxic cells. Therefore, assays that allow for the simultaneous measurement of several parameters may be more advantageous for clinical monitoring. In this respect, multifactor flow cytometry-based assays are a valid addition to the currently available immunologic monitoring assays. Use of these assays will enable detection and enumeration of tumor-specific cytotoxic T lymphocytes and their specific effector functions and any correlations with clinical responses. Comprehensive, multifactor analysis of effector cell responses after vaccination may help to detect factors that determine the success or failure of a vaccine and its immunological potency.
Collapse
Affiliation(s)
- Liubov Zaritskaya
- Applied and Developmental Research Support Program, SAIC-Frederick,
Inc., National Cancer Institute at Frederick, Frederick, MD, USA
| | - Michael R Shurin
- Departments of Pathology and Immunology, University of Pittsburgh
Medical Center, Pittsburgh, PA, USA
| | - Thomas J Sayers
- Cancer and Inflammation Program, SAIC-Frederick, Inc., National
Cancer Institute at Frederick, Frederick, MD, USA
| | - Anatoli M Malyguine
- Applied and Developmental Research Support Program, SAIC-Frederick,
Inc., National Cancer Institute at Frederick, Frederick, MD, USA
| |
Collapse
|
90
|
Libanova R, Ebensen T, Schulze K, Bruhn D, Nörder M, Yevsa T, Morr M, Guzmán CA. The member of the cyclic di-nucleotide family bis-(3', 5')-cyclic dimeric inosine monophosphate exerts potent activity as mucosal adjuvant. Vaccine 2010; 28:2249-2258. [PMID: 20060510 DOI: 10.1016/j.vaccine.2009.12.045] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2009] [Revised: 12/11/2009] [Accepted: 12/20/2009] [Indexed: 01/01/2023]
Abstract
Here we demonstrated that bis-(3',5')-cyclic dimeric inosine monophosphate (c-di-IMP) exhibits potent adjuvant properties. BALB/c or C57BL/6 mice were immunized with the model antigens beta-galactosidase (beta-Gal) or Ovalbumin (OVA) alone or co-administered with c-di-IMP by the intranasal route. Animals receiving c-di-IMP showed significantly higher anti-beta-Gal or OVA immunoglobulin G titres (IgG) in sera than those vaccinated with beta-Gal or OVA alone. Furthermore, strong local immune responses were also detectable in different mucosal territories, as shown by the high levels of beta-Gal-specific secretory IgA (sIgA). The analysis of the antigen-specific IgG isotypes in sera, together with the profiles of the cytokines and chemokines secreted by lymphocytes from vaccinated animals showed that the use of c-di-IMP resulted in stimulation of a mixed T(H)1/T(H)2/T(H)17 response. Mucosal immunization of C57BL/6 mice with OVA using c-di-IMP as adjuvant also led to the stimulation of strong in vivo CTL responses (i.e., 60% of antigen-specific lysis) [corrected].Our results demonstrated that the novel compound c-di-IMP exhibits strong adjuvant properties when co-administered with an antigen by the mucosal route, thereby representing a promising candidate adjuvant for the development of mucosal vaccination strategies.
Collapse
Affiliation(s)
- Rimma Libanova
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre of Infection Research, Inhoffenstrasse 7, D-38124 Braunschweig, Germany
| | - Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre of Infection Research, Inhoffenstrasse 7, D-38124 Braunschweig, Germany.
| | - Kai Schulze
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre of Infection Research, Inhoffenstrasse 7, D-38124 Braunschweig, Germany
| | - Daniela Bruhn
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre of Infection Research, Inhoffenstrasse 7, D-38124 Braunschweig, Germany
| | - Miriam Nörder
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre of Infection Research, Inhoffenstrasse 7, D-38124 Braunschweig, Germany
| | - Tetyana Yevsa
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre of Infection Research, Inhoffenstrasse 7, D-38124 Braunschweig, Germany
| | - Michael Morr
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre of Infection Research, Inhoffenstrasse 7, D-38124 Braunschweig, Germany
| | - Carlos A Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre of Infection Research, Inhoffenstrasse 7, D-38124 Braunschweig, Germany
| |
Collapse
|
91
|
Abstract
Natural killer (NK) cells were discovered in the 1970 s and named after their naturally occurring cytotoxic activity against tumor cells. It has recently become clear that NK cells are not just killers and that malignancy is unlikely to be the selective pressure driving the evolution of NK cells. Indeed, NK cells secrete a host of cytokines and chemokines that contribute to tissue remodeling at the feto-maternal interface and to both innate and adaptive immunity during infection. Moreover, in certain conditions, they cannot deliver functions cell autonomously, as they require priming from other cells, namely dendritic cells. Nevertheless, natural cytotoxicity is still considered an important parameter used to evaluate NK cell biology, both in the clinic and in the research lab. In this chapter we describe a simple method to quantify spontaneous NK cell cytotoxicity in vivo.
Collapse
Affiliation(s)
- Aurore Saudemont
- Lymphocyte Signalling and Development, The Babraham Institute, Cambridge, UK
| | | | | |
Collapse
|
92
|
Robinson MJ, Ronchese F, Miller JH, La Flamme AC. Paclitaxel inhibits killing by murine cytotoxic T lymphocytes in vivo
but not in vitro. Immunol Cell Biol 2009; 88:291-6. [DOI: 10.1038/icb.2009.96] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Marcus James Robinson
- School of Biological Sciences, Victoria University of Wellington; Wellington New Zealand
| | - Franca Ronchese
- Malaghan Institute of Medical Research; Wellington New Zealand
| | - John H Miller
- School of Biological Sciences, Victoria University of Wellington; Wellington New Zealand
- Centre for Biodiscovery, Victoria University of Wellington; Wellington New Zealand
| | - Anne Camille La Flamme
- School of Biological Sciences, Victoria University of Wellington; Wellington New Zealand
- Centre for Biodiscovery, Victoria University of Wellington; Wellington New Zealand
| |
Collapse
|
93
|
Tissue-specific differences in PD-1 and PD-L1 expression during chronic viral infection: implications for CD8 T-cell exhaustion. J Virol 2009; 84:2078-89. [PMID: 19955307 DOI: 10.1128/jvi.01579-09] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The PD-1/PD-L pathway plays a major role in regulating T-cell exhaustion during chronic viral infections in animal models, as well as in humans, and blockade of this pathway can revive exhausted CD8(+) T cells. We examined the expression of PD-1 and its ligands, PD-L1 and PD-L2, in multiple tissues during the course of chronic viral infection and determined how the amount of PD-1 expressed, as well as the anatomical location, influenced the function of exhausted CD8 T cells. The amount of PD-1 on exhausted CD8 T cells from different anatomical locations did not always correlate with infectious virus but did reflect viral antigen in some tissues. Moreover, lower expression of PD-L1 in some locations, such as the bone marrow, favored the survival of PD-1(Hi) exhausted CD8 T cells, suggesting that some anatomical sites might provide a survival niche for subpopulations of exhausted CD8 T cells. Tissue-specific differences in the function of exhausted CD8 T cells were also observed. However, while cytokine production did not strictly correlate with the amount of PD-1 expressed by exhausted CD8 T cells from different tissues, the ability to degranulate and kill were tightly linked to PD-1 expression regardless of the anatomical location. These observations have implications for human chronic infections and for therapeutic interventions based on blockade of the PD-1 pathway.
Collapse
|
94
|
Sorensen MR, Holst PJ, Pircher H, Christensen JP, Thomsen AR. Vaccination with an adenoviral vector encoding the tumor antigen directly linked to invariant chain induces potent CD4+T-cell-independent CD8+T-cell-mediated tumor control. Eur J Immunol 2009; 39:2725-36. [DOI: 10.1002/eji.200939543] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
95
|
Development of highly sensitive Bicistronic vector based non-radioactive antigen-specific cytotoxicity assay. J Immunol Methods 2009; 349:28-37. [DOI: 10.1016/j.jim.2009.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2009] [Revised: 08/01/2009] [Accepted: 08/04/2009] [Indexed: 01/12/2023]
|
96
|
Kochenderfer JN, Feldman SA, Zhao Y, Xu H, Black MA, Morgan RA, Wilson WH, Rosenberg SA. Construction and preclinical evaluation of an anti-CD19 chimeric antigen receptor. J Immunother 2009; 32:689-702. [PMID: 19561539 PMCID: PMC2747302 DOI: 10.1097/cji.0b013e3181ac6138] [Citation(s) in RCA: 329] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
T cells can be engineered to express the genes of chimeric antigen receptors (CARs) that recognize tumor-associated antigens. We constructed and compared 2 CARs that contained a single chain variable region moiety that recognized CD19. One CAR contained the signaling moiety of the 4-1BB molecule and the other did not. We selected the CAR that did not contain the 4-1BB moiety for further preclinical development. We demonstrated that gammaretroviruses encoding this receptor could transduce human T cells. Anti-CD19-CAR-transduced CD8+ and CD4+ T cells produced interferon-gamma and interleukin-2 specifically in response to CD19+ target cells. The transduced T cells specifically killed primary chronic lymphocytic leukemia (CLL) cells. We transduced T cells from CLL patients that had been previously treated with chemotherapy. We induced these T cells to proliferate sufficiently to provide enough cells for clinical adoptive T cell transfer with a protocol consisting of an initial stimulation with an anti-CD3 monoclonal antibody (OKT3) before transduction followed by a second OKT3 stimulation 7 days after transduction. This protocol was successfully adapted for use in CLL patients with high peripheral blood leukemia cell counts by depleting CD19+ cells before the initial OKT3 stimulation. In preparation for a clinical trial that will enroll patients with advanced B cell malignancies, we generated a producer cell clone that produces retroviruses encoding the anti-CD19 CAR, and we produced sufficient retroviral supernatant for the proposed clinical trial under good manufacturing practice conditions.
Collapse
MESH Headings
- Antigens, CD19/genetics
- Antigens, CD19/immunology
- Antigens, CD19/metabolism
- CD4-Positive T-Lymphocytes/cytology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Line, Tumor
- Cytotoxicity Tests, Immunologic
- Cytotoxicity, Immunologic/immunology
- Drug Evaluation, Preclinical
- Enzyme-Linked Immunosorbent Assay
- Gammaretrovirus/genetics
- Genetic Vectors/genetics
- Humans
- Immunotherapy/methods
- Interferon-gamma/metabolism
- Interleukin-2/metabolism
- K562 Cells
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Muromonab-CD3/immunology
- Receptors, Antigen/genetics
- Receptors, Antigen/immunology
- Receptors, Nerve Growth Factor/genetics
- Receptors, Nerve Growth Factor/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/metabolism
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Transduction, Genetic
Collapse
Affiliation(s)
- James N Kochenderfer
- Surgery Branch of the National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | |
Collapse
|
97
|
Giagulli C, Noerder M, Avolio M, Becker PD, Fiorentini S, Guzman CA, Caruso A. Pidotimod promotes functional maturation of dendritic cells and displays adjuvant properties at the nasal mucosa level. Int Immunopharmacol 2009; 9:1366-73. [PMID: 19712757 DOI: 10.1016/j.intimp.2009.08.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2009] [Revised: 08/07/2009] [Accepted: 08/10/2009] [Indexed: 10/20/2022]
Abstract
Mucosal dendritic cells (DCs) are very important in the process of antigen presentation to T cells, playing a key role in the induction of primary and secondary immune responses. Pidotimod is a synthetic substance capable of modulating immune cell functions, but the effect of pidotimod on human DCs has not been investigated yet. Here we demonstrate the ability of pidotimod to induce DC maturation and up-regulate the expression of HLA-DR and co-stimulatory molecules CD83 and CD86, which are fundamental for communication with adaptative immunity cells. Pidotimod also stimulated DCs to release high amounts of pro-inflammatory molecules such as MCP-1 and TNF-alpha cytokines and to drive T cell proliferation and differentiation towards a Th1 phenotype. Moreover, we demonstrate that pidotimod in vivo promotes strong and specific humoral and cellular immune response when co-administered intranasally with a model antigen. Taken together our data suggest the possibility to use pidotimod as adjuvant molecule to facilitate the activation of the innate immune system as well as to promote an effective mucosal and systemic immune response.
Collapse
Affiliation(s)
- Cinzia Giagulli
- Department of Experimental and Applied Medicine, Section of Microbiology, University of Brescia, Medical School, Brescia, Italy
| | | | | | | | | | | | | |
Collapse
|
98
|
Shin H, Blackburn SD, Intlekofer AM, Kao C, Angelosanto JM, Reiner SL, Wherry EJ. A role for the transcriptional repressor Blimp-1 in CD8(+) T cell exhaustion during chronic viral infection. Immunity 2009; 31:309-20. [PMID: 19664943 PMCID: PMC2747257 DOI: 10.1016/j.immuni.2009.06.019] [Citation(s) in RCA: 392] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2009] [Revised: 06/15/2009] [Accepted: 06/29/2009] [Indexed: 01/01/2023]
Abstract
T cell exhaustion is common during chronic infections and can prevent optimal immunity. Although recent studies have demonstrated the importance of inhibitory receptors and other pathways in T cell exhaustion, the underlying transcriptional mechanisms are unknown. Here, we define a role for the transcription factor Blimp-1 in CD8(+) T cell exhaustion during chronic viral infection. Blimp-1 repressed key aspects of normal memory CD8(+) T cell differentiation and promoted high expression of inhibitory receptors during chronic infection. These cardinal features of CD8(+) T cell exhaustion were corrected by conditionally deleting Blimp-1. Although high expression of Blimp-1 fostered aspects of CD8(+) T cell exhaustion, haploinsufficiency indicated that moderate Blimp-1 expression sustained some effector function during chronic viral infection. Thus, we identify Blimp-1 as a transcriptional regulator of CD8(+) T cell exhaustion during chronic viral infection and propose that Blimp-1 acts as a transcriptional rheostat balancing effector function and T cell exhaustion.
Collapse
MESH Headings
- Acute Disease
- Animals
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Antigens, Surface/immunology
- Antigens, Surface/metabolism
- Apoptosis Regulatory Proteins/immunology
- Apoptosis Regulatory Proteins/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/virology
- Cell Differentiation/immunology
- Chronic Disease
- Cytotoxicity, Immunologic/immunology
- GPI-Linked Proteins
- Granzymes/immunology
- Granzymes/metabolism
- Immunologic Memory/immunology
- Lymphocytic Choriomeningitis/immunology
- Lymphocytic Choriomeningitis/virology
- Lymphocytic choriomeningitis virus/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Positive Regulatory Domain I-Binding Factor 1
- Programmed Cell Death 1 Receptor
- Receptors, Immunologic/immunology
- Receptors, Immunologic/metabolism
- Signaling Lymphocytic Activation Molecule Family
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Virus Diseases/genetics
- Virus Diseases/immunology
- Lymphocyte Activation Gene 3 Protein
Collapse
Affiliation(s)
- Haina Shin
- Immunology Program and Wistar Vaccine Center, The Wistar Institute, Philadelphia, PA 19104 USA
| | - Shawn D. Blackburn
- Immunology Program and Wistar Vaccine Center, The Wistar Institute, Philadelphia, PA 19104 USA
| | - Andrew M. Intlekofer
- Abramson Family Cancer Research Institute, Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - Charlly Kao
- Immunology Program and Wistar Vaccine Center, The Wistar Institute, Philadelphia, PA 19104 USA
| | - Jill M. Angelosanto
- Immunology Program and Wistar Vaccine Center, The Wistar Institute, Philadelphia, PA 19104 USA
| | - Steven L. Reiner
- Abramson Family Cancer Research Institute, Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104 USA
| | - E. John Wherry
- Immunology Program and Wistar Vaccine Center, The Wistar Institute, Philadelphia, PA 19104 USA
| |
Collapse
|
99
|
Tripp CH, Sparber F, Hermans IF, Romani N, Stoitzner P. Glycolipids Injected into the Skin Are Presented to NKT Cells in the Draining Lymph Node Independently of Migratory Skin Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2009; 182:7644-54. [DOI: 10.4049/jimmunol.0900134] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
100
|
Palmowski MJ, Parker M, Choudhuri K, Chiu C, Callan MFC, van der Merwe PA, Cerundolo V, Gould KG. A single-chain H-2Db molecule presenting an influenza virus nucleoprotein epitope shows enhanced ability at stimulating CD8+ T cell responses in vivo. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 182:4565-71. [PMID: 19342630 DOI: 10.4049/jimmunol.0803893] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have generated a construct encoding a single-chain H-2D(b) mouse MHC class I molecule in which an influenza virus nucleoprotein (NP) epitope, amino acid sequence ASNENMDAM, is fused to mouse beta(2)-microglobulin and the D(b) H chain via flexible linker sequences. This single-chain trimer (SCT) was efficiently expressed at the cell surface independently of TAP and endogenous beta(2)-microglobulin, and it was recognized directly and efficiently by specific T cells in vitro. A recombinant vaccinia virus encoding the D(b) NP SCT primed a CD8(+) T cell response in C57BL/6 mice 4-fold greater than an equivalent virus expressing the NP epitope as a minigene, as shown by tetramer staining, whether or not the minigene was directed into the endoplasmic reticulum by a signal sequence. This response was functional as shown by in vivo lysis assays with peptide-pulsed target cells, and it was greatly expanded following secondary challenge in vivo with influenza virus. The SCT was also significantly more immunostimulatory for CD8(+) cells than the NP minigene in adoptive transfer experiments using F5 TCR transgenic spleen cells, in which the magnitude of the T cell response was much greater. Our results extend previous DNA vaccination studies using SCTs, which demonstrated that such molecules are capable of generating functional CD8(+) T cell responses. We have shown that class I SCTs are more immunogenic than even preprocessed Ag in the form of an epitope minigene, and they therefore should be considered for use when the generation of optimal CD8(+) T cell responses is required.
Collapse
Affiliation(s)
- Michael J Palmowski
- Tumour Immunology Group, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|