1
|
Kajihara N, Ge Y, Seino KI. Blocking of oestrogen signals improves anti-tumour effect regardless of oestrogen receptor alpha expression in cancer cells. Br J Cancer 2023; 129:935-946. [PMID: 37537255 PMCID: PMC10491758 DOI: 10.1038/s41416-023-02381-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/10/2023] [Accepted: 07/25/2023] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND Anti-oestrogenic therapy has been used for breast cancer patients with oestrogen susceptibility cancer cells. However, little has been known about its potential role for immune cell biology within TME, particularly in cancer patients without oestrogen sensitivity of tumour cells. Therefore, we aimed to study the effect of oestrogen on immunity within TME. METHODS Using a clinical dataset, immune cells of humans and mice, female mice with and without ovaries, and several murine ERα-negative cancer cell lines, we evaluated the effect of oestrogen on immunity in TME. RESULTS Clinical data analysis suggested oestrogen's suppressive efficacy against CTLs. Additionally, in vitro and in vivo experiments revealed intra-tumoural CTLs' direct repressive action by oestrogen in both mice and humans; blockade of oestrogen signals cancelled its immunosuppression resulting in tumour growth reduction in vivo. Most notably, immunotherapy (immune checkpoint inhibitor; ICI) combined with anti-oestrogenic therapy exhibited a dramatic anti-tumour effect. CONCLUSIONS This study provides novel insights into how oestrogen contributes to tumour progression and a therapeutic rationale for blocking oestrogen signalling to boost the anti-tumour effect of ICI, regardless of tumour cells' ERα expression.
Collapse
Affiliation(s)
- Nabeel Kajihara
- Division of Immunobiology, Graduate School of Medicine, Institute for Genetic Medicine, Hokkaido University, Kita-15 Nishi-7, Sapporo, 060-0815, Japan
| | - Yunqi Ge
- Division of Immunobiology, Graduate School of Medicine, Institute for Genetic Medicine, Hokkaido University, Kita-15 Nishi-7, Sapporo, 060-0815, Japan
| | - Ken-Ichiro Seino
- Division of Immunobiology, Graduate School of Medicine, Institute for Genetic Medicine, Hokkaido University, Kita-15 Nishi-7, Sapporo, 060-0815, Japan.
| |
Collapse
|
2
|
Leitner J, Egerer R, Waidhofer-Söllner P, Grabmeier-Pfistershammer K, Steinberger P. FcγR requirements and costimulatory capacity of Urelumab, Utomilumab, and Varlilumab. Front Immunol 2023; 14:1208631. [PMID: 37575254 PMCID: PMC10413977 DOI: 10.3389/fimmu.2023.1208631] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/28/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction Targeting costimulatory receptors of the tumor necrosis factor receptor (TNFR) superfamily with agonistic antibodies is a promising approach in cancer immuno therapy. It is known that their efficacy strongly depends on FcγR cross-linking. Methods In this study, we made use of a Jurkat-based reporter platform to analyze the influence of individual FcγRs on the costimulatory activity of the 41BB agonists, Urelumab and Utomilumab, and the CD27 agonist, Varlilumab. Results We found that Urelumab (IgG4) can activate 41BB-NFκB signaling without FcγR cross-linking, but the presence of the FcγRs (CD32A, CD32B, CD64) augments the agonistic activity of Urelumab. The human IgG2 antibody Utomilumab exerts agonistic function only when crosslinked via CD32A and CD32B. The human IgG1 antibody Varlilumab showed strong agonistic activity with all FcγRs tested. In addition, we analyzed the costimulatory effects of Urelumab, Utomilumab, and Varlilumab in primary human peripheral blood mononuclear cells (PBMCs). Interestingly, we observed a very weak capacity of Varlilumab to enhance cytokine production and proliferation of CD4 and CD8 T cells. In the presence of Varlilumab the percentage of annexin V positive T cells was increased, indicating that this antibody mediated FcγR-dependent cytotoxic effects. Conclusion Collectively, our data underscore the importance to perform studies in reductionist systems as well as in primary PBMC samples to get a comprehensive understanding of the activity of costimulation agonists.
Collapse
Affiliation(s)
- Judith Leitner
- Division of Immune Receptors and T Cell Activation, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Ricarda Egerer
- Division of Immune Receptors and T Cell Activation, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Petra Waidhofer-Söllner
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | | | - Peter Steinberger
- Division of Immune Receptors and T Cell Activation, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
3
|
Dib M, Le Corre N, Ortiz C, García D, Ferrés M, Martinez-Valdebenito C, Ruiz-Tagle C, Ojeda MJ, Espinoza MA, Jara A, Arab JP, Rabagliati R, Vizcaya C, Ceballos ME, Sarmiento M, Mondaca S, Viñuela M, Pastore A, Szwarcfiter V, Galdames E, Barrera A, Castro P, Gálvez NMS, Soto JA, Bueno SM, Kalergis AM, Nervi B, Balcells ME. SARS-CoV-2 vaccine booster in solid organ transplant recipients previously immunised with inactivated versus mRNA vaccines: A prospective cohort study. LANCET REGIONAL HEALTH. AMERICAS 2022; 16:100371. [PMID: 36185969 PMCID: PMC9503242 DOI: 10.1016/j.lana.2022.100371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Background Solid-organ transplant (SOT) recipients have worse COVID-19 outcomes than general population and effective immunisation in these patients is essential but more difficult to reach. We aimed to determine the immunogenicity of an mRNA SARS-CoV-2 vaccine booster in SOT recipients previously immunised with either inactivated or homologous SARS-CoV-2 mRNA vaccine. Methods Prospective cohort study of SOT recipients under medical care at Red de Salud UC-CHRISTUS, Chile, previously vaccinated with either CoronaVac or BNT162b2. All participants received a BNT162b2 vaccine booster. The primary study end point was anti-SARS-CoV-2 total IgG antibodies (TAb) seropositivity at 8-12 weeks (56-84 days) post booster. Secondary end points included neutralising antibodies (NAb) and specific T-cell responses. Findings A total of 140 (50% kidney, 38% liver, 6% heart) SOT recipients (mean age 54 [13.6] years; 64 [46%] women) were included. Of them, 62 had homologous (three doses of BNT162b2) and 78 heterologous vaccine schedules (two doses of CoronaVac followed by BNT162b2 booster). Boosters were received at a median of 21.3 weeks after primary vaccination. The proportion achieving TAb seropositivity (82.3% vs 65.4%, P = 0.035) and NAb positivity (77.4% vs 55.1%, P = 0.007) were higher for the homologous versus the heterologous group. On the other hand, the number of IFN-γ and IL-2 secreting SARS-CoV-2-specific T-cells did not differ significantly between groups. Interpretation This cohort study shows that homologous mRNA vaccine priming plus boosting in SOT recipients, reaches a significantly higher humoral immune response than inactivated SARS-CoV-2 vaccine priming followed by heterologous mRNA booster. Funding School of Medicine, UC-Chile and ANID.ClinicalTrials.gov ID: NCT05124509.
Collapse
Affiliation(s)
- Martín Dib
- Instituto de Trasplantes, Red de Salud UC CHRISTUS, Chile
- Departamento de Cirugía Digestiva, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicole Le Corre
- Laboratorio de Infectología y Virología Molecular, Red de Salud UC CHRISTUS, Chile
- Departamento de Enfermedades Infecciosas e Inmunología Pediátrica, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina Ortiz
- Departamento de Cirugía Digestiva, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniel García
- Departamento de Cirugía Digestiva, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Equipo de Cirugía Digestiva, Servicio de Cirugía, Hospital Dr Eduardo Pereira, Valparaíso, Chile
| | - Marcela Ferrés
- Laboratorio de Infectología y Virología Molecular, Red de Salud UC CHRISTUS, Chile
- Departamento de Enfermedades Infecciosas e Inmunología Pediátrica, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Constanza Martinez-Valdebenito
- Laboratorio de Infectología y Virología Molecular, Red de Salud UC CHRISTUS, Chile
- Departamento de Enfermedades Infecciosas e Inmunología Pediátrica, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cinthya Ruiz-Tagle
- Departamento de Enfermedades Infecciosas del Adulto, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María José Ojeda
- Departamento de Enfermedades Infecciosas del Adulto, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Manuel A. Espinoza
- Departamento de Salud Pública, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Aquiles Jara
- Departamento de Nefrología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Pablo Arab
- Departamento de Gastroenterología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ricardo Rabagliati
- Departamento de Enfermedades Infecciosas del Adulto, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cecilia Vizcaya
- Departamento de Enfermedades Infecciosas e Inmunología Pediátrica, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María Elena Ceballos
- Departamento de Enfermedades Infecciosas del Adulto, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mauricio Sarmiento
- Departamento de Hematología y Oncología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sebastián Mondaca
- Departamento de Hematología y Oncología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Macarena Viñuela
- Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Antonia Pastore
- Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Vania Szwarcfiter
- Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Aldo Barrera
- Laboratorio de Infectología y Virología Molecular, Red de Salud UC CHRISTUS, Chile
- Departamento de Enfermedades Infecciosas e Inmunología Pediátrica, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo Castro
- División de Enfermedades Cardiovasculares, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolás MS Gálvez
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile. Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge A. Soto
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile. Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile. Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile. Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bruno Nervi
- Departamento de Hematología y Oncología, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - M. Elvira Balcells
- Departamento de Enfermedades Infecciosas del Adulto, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
4
|
Rani L, Kumar A, Karhade J, Pandey G, Guha A, Mishra GC, Wani MR. IL-3 regulates the differentiation of pathogenic Th17 cells. Eur J Immunol 2022; 52:1842-1858. [PMID: 36074916 DOI: 10.1002/eji.202149674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 07/26/2022] [Accepted: 09/06/2022] [Indexed: 11/06/2022]
Abstract
IL-17 producing Th17 cells play an important role in pathogenesis of rheumatoid arthritis (RA). Aberrant immune activation due to imbalance between Th17 and regulatory T (Treg) cells is associated with development of RA and other autoimmune diseases. Targeting pathogenic Th17 cells and their associated molecules is emerging as a promising strategy to treat and reverse RA. Here, we demonstrate that IL-3 inhibits the differentiation of Th17 cells and promotes the development of Treg cells in IL-2-dependent manner. In IL-2 knockout mice, we observed that IL-3 has no effect on differentiation of both Th17 and Treg cells. In addition, IL-3 decreases pathogenic IL-17A+ TNF-α+ , IL-17A+ IFN-γ+ and IL-23R+ Th17 cells, secretion of GM-CSF and IFN-γ, and osteoclastogenesis when presented in the culture together with Th-17 polarizing cytokines. Mechanistically, IL-3 regulates the development of Th17 cells through inhibition of STAT3 phosphorylation. IL-3 treatment significantly decreases the pathogenic Th17 cell responses and arthritic scores in mouse model of RA. Importantly, IL-3 inhibits differentiation of human Th17 cells. Thus, our results suggest a novel therapeutic role of IL-3 in regulation of Th17 cell-mediated pathophysiology of RA. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Lekha Rani
- Bone and Cartilage Research Laboratory, National Centre for Cell Science, Pune, 411007, India
| | - Anil Kumar
- Bone and Cartilage Research Laboratory, National Centre for Cell Science, Pune, 411007, India
| | - Juilee Karhade
- Bone and Cartilage Research Laboratory, National Centre for Cell Science, Pune, 411007, India
| | - Garima Pandey
- Bone and Cartilage Research Laboratory, National Centre for Cell Science, Pune, 411007, India
| | - Adrita Guha
- Bone and Cartilage Research Laboratory, National Centre for Cell Science, Pune, 411007, India
| | - Gyan C Mishra
- Bone and Cartilage Research Laboratory, National Centre for Cell Science, Pune, 411007, India
| | - Mohan R Wani
- Bone and Cartilage Research Laboratory, National Centre for Cell Science, Pune, 411007, India
| |
Collapse
|
5
|
Immunogenicity of Varicella Zoster Virus DNA Vaccines Encoding Glycoprotein E and Immediate Early Protein 63 in Mice. Viruses 2022; 14:v14061214. [PMID: 35746685 PMCID: PMC9230688 DOI: 10.3390/v14061214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 02/05/2023] Open
Abstract
Herpes zoster (HZ) is caused by the reactivation of latent varicella-zoster virus (VZV) from the sensory ganglia due to aging or immunosuppression. Glycoprotein E (gE) is a widely used vaccine antigen for specific humoral and cellular immune responses. Immediate early protein 63 (IE63) is expressed during latency, suggesting that it is a potential antigen against HZ reactivation. In this study, HZ DNA vaccines encoding gE, IE63, IE63-2A-gE (where 2A is a self-cleaving sequence), or IE63-linker-gE were developed and investigated for immunogenicity in mice. The results showed that each HZ DNA vaccine induced VZV-specific antibody production. The neutralizing antibody titer elicited by IE63-2A-gE was comparable to that elicited by gE or live attenuated HZ vaccine (LAV). IE63-2A-gE-induced gE or IE63-specific INF-γ+ T cell frequencies in splenocytes were comparable to those of LAV. Furthermore, IE63-2A-gE, gE, or IE63 led to a significant increase in IFN-γ (IE63 stimulation) and IL-2 (gE stimulation) secretion compared to LAV, showing a Th1-biased immune response. Moreover, IE63-2A-gE and gE induced cytotoxic activity of CD8+ T cells compared to that of LAV. This study elucidates that the IE63-2A-gE DNA vaccine can induce both humoral and cell-mediated immune responses, which provides a candidate for the development of an HZ vaccine.
Collapse
|
6
|
The generation and application of antigen-specific T cell therapies for cancer and viral-associated disease. Mol Ther 2022; 30:2130-2152. [PMID: 35149193 PMCID: PMC9171249 DOI: 10.1016/j.ymthe.2022.02.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/27/2021] [Accepted: 02/03/2022] [Indexed: 11/23/2022] Open
Abstract
Immunotherapy with antigen-specific T cells is a promising, targeted therapeutic option for patients with cancer as well as for immunocompromised patients with virus infections. In this review, we characterize and compare current manufacturing protocols for the generation of T cells specific to viral and non-viral tumor-associated antigens. Specifically, we discuss: (1) the different methodologies to expand virus-specific T cell and non-viral tumor-associated antigen-specific T cell products, (2) an overview of the immunological principles involved when developing such manufacturing protocols, and (3) proposed standardized methodologies for the generation of polyclonal, polyfunctional antigen-specific T cells irrespective of donor source. Ex vivo expanded cells have been safely administered to treat numerous patients with virus-associated malignancies, hematologic malignancies, and solid tumors. Hence, we have performed a comprehensive review of the clinical trial results evaluating the safety, feasibility, and efficacy of these products in the clinic. In summary, this review seeks to provide new insights regarding antigen-specific T cell technology to benefit a rapidly expanding T cell therapy field.
Collapse
|
7
|
Mu P, Huo J, Sun M, Chen X, Ao J. Identification and expression analysis of IL-2 receptors in large yellow croaker (Larimichthys crocea). FISH AND SHELLFISH IMMUNOLOGY REPORTS 2021; 2:100008. [DOI: 10.1016/j.fsirep.2021.100008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/03/2021] [Accepted: 03/11/2021] [Indexed: 01/06/2023] Open
|
8
|
Samojlik MM, Stabler CL. Designing biomaterials for the modulation of allogeneic and autoimmune responses to cellular implants in Type 1 Diabetes. Acta Biomater 2021; 133:87-101. [PMID: 34102338 PMCID: PMC9148663 DOI: 10.1016/j.actbio.2021.05.039] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/05/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022]
Abstract
The effective suppression of adaptive immune responses is essential for the success of allogeneic cell therapies. In islet transplantation for Type 1 Diabetes, pre-existing autoimmunity provides an additional hurdle, as memory autoimmune T cells mediate both an autoantigen-specific attack on the donor beta cells and an alloantigen-specific attack on the donor graft cells. Immunosuppressive agents used for islet transplantation are generally successful in suppressing alloimmune responses, but dramatically hinder the widespread adoption of this therapeutic approach and fail to control memory T cell populations, which leaves the graft vulnerable to destruction. In this review, we highlight the capacity of biomaterials to provide local and nuanced instruction to suppress or alter immune pathways activated in response to an allogeneic islet transplant. Biomaterial immunoisolation is a common approach employed to block direct antigen recognition and downstream cell-mediated graft destruction; however, immunoisolation alone still permits shed donor antigens to escape into the host environment, resulting in indirect antigen recognition, immune cell activation, and the creation of a toxic graft site. Designing materials to decrease antigen escape, improve cell viability, and increase material compatibility are all approaches that can decrease the local release of antigen and danger signals into the implant microenvironment. Implant materials can be further enhanced through the local delivery of anti-inflammatory, suppressive, chemotactic, and/or tolerogenic agents, which serve to control both the innate and adaptive immune responses to the implant with a benefit of reduced systemic effects. Lessons learned from understanding how to manipulate allogeneic and autogenic immune responses to pancreatic islets can also be applied to other cell therapies to improve their efficacy and duration. STATEMENT OF SIGNIFICANCE: This review explores key immunologic concepts and critical pathways mediating graft rejection in Type 1 Diabetes, which can instruct the future purposeful design of immunomodulatory biomaterials for cell therapy. A summary of immunological pathways initiated following cellular implantation, as well as current systemic immunomodulatory agents used, is provided. We then outline the potential of biomaterials to modulate these responses. The capacity of polymeric encapsulation to block some powerful rejection pathways is covered. We also highlight the role of cellular health and biocompatibility in mitigating immune responses. Finally, we review the use of bioactive materials to proactively modulate local immune responses, focusing on key concepts of anti-inflammatory, suppressive, and tolerogenic agents.
Collapse
Affiliation(s)
- Magdalena M Samojlik
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Cherie L Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA; University of Florida Diabetes Institute, Gainesville, FL, USA; Graduate Program in Biomedical Sciences, College of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
9
|
Kim YH, Bang YJ, Park HJ, Li Ko H, Park SI, Hwang KA, Kim H, Nam JH. Inactivated influenza vaccine formulated with single-stranded RNA-based adjuvant confers mucosal immunity and cross-protection against influenza virus infection. Vaccine 2020; 38:6141-6152. [DOI: 10.1016/j.vaccine.2020.07.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 06/20/2020] [Accepted: 07/12/2020] [Indexed: 01/31/2023]
|
10
|
Amani MF, Rolig AS, Redmond WL. Intracellular Galectin-3 Is Essential for OX40-Mediated Memory CD8+ T Cell Development. THE JOURNAL OF IMMUNOLOGY 2020; 205:1857-1866. [DOI: 10.4049/jimmunol.1901052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 08/01/2020] [Indexed: 11/19/2022]
|
11
|
Lee SJ, Park HJ, Ko HL, Lee JE, Lee HJ, Kim H, Nam JH. Evaluation of glycoprotein E subunit and live attenuated varicella-zoster virus vaccines formulated with a single-strand RNA-based adjuvant. IMMUNITY INFLAMMATION AND DISEASE 2020; 8:216-227. [PMID: 32167678 PMCID: PMC7212201 DOI: 10.1002/iid3.297] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 02/25/2020] [Accepted: 02/27/2020] [Indexed: 12/24/2022]
Abstract
Introduction Varicella‐zoster virus (VZV), a human alphaherpesvirus 3, elicits both chickenpox and shingles and/or postherpetic neuralgia. A live attenuated vaccine (LAV) and glycoprotein E (gE) subunit vaccine were developed to prevent VZV‐induced diseases. We recently reported that single‐strand RNA (ssRNA) based on the intergenic region of the internal ribosome entry site of cricket paralysis virus (CrPV) is an effective adjuvant for protein‐based and virus‐like particle‐based vaccines. Here, Chinese hamster ovary expression system and an LAV from Oka/SK strains. Methods We appraised the adjuvant effect of the same CrPV ssRNA encoding the gE gene formulated in the two vaccines using VZV‐primed C57BL/6 mice and guinea pigs. Humoral immunity and cell‐mediated immunity were assessed by enzyme‐linked immunosorbent assay (ELISA) and ELISPOT in gE subunit vaccine and by ELISA and fluorescent antibody to membrane antigen in LAV. Results The gE subunit vaccine‐induced gE‐specific antibodies and CD4+ T‐cell responses (indicated by interferon‐γ [IFN‐γ] and interleukin‐2 secretion) in the ssRNA‐based adjuvant containing the VZV gE gene. Therefore, an ssRNA adjuvant combined with gE antigen can trigger the innate immune response and induce an adaptive immune response to ultimately activate humoral and cell‐mediated responses. VZV LAV could also induce VZV‐specific antibodies and IFN‐γ stimulated by LAV, whereas the effect of ssRNA as a vaccine adjuvant could not be confirmed. However, the ssRNA adjuvant increased VZV‐specific neutralizing antibody response. Conclusions Taken together, these results highlight that the gE subunit vaccine and LAV developed in this study can be functional VZV vaccines, and ssRNAs appear to function better as adjuvants in a subunit vaccine than in an LAV.
Collapse
Affiliation(s)
- Su Jeen Lee
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea.,Department of R&D, SK Bioscience, Pangyoro, Bundang-gu, Republic of Korea
| | - Hyo-Jung Park
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Hae Li Ko
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Jung Eun Lee
- Department of R&D, SK Bioscience, Pangyoro, Bundang-gu, Republic of Korea
| | - Hyun Joo Lee
- Department of R&D, SK Bioscience, Pangyoro, Bundang-gu, Republic of Korea
| | - Hun Kim
- Department of R&D, SK Bioscience, Pangyoro, Bundang-gu, Republic of Korea
| | - Jae-Hwan Nam
- Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
| |
Collapse
|
12
|
Zeng YQ, Liu XS, Wu S, Zou C, Xie Q, Xu SM, Jin XW, Li W, Zhou A, Dai Z. Kaempferol Promotes Transplant Tolerance by Sustaining CD4+FoxP3+ Regulatory T Cells in the Presence of Calcineurin Inhibitor. Am J Transplant 2015; 15:1782-92. [PMID: 25808405 DOI: 10.1111/ajt.13261] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 02/01/2015] [Accepted: 02/05/2015] [Indexed: 01/25/2023]
Abstract
Calcineurin inhibitor cyclosporine is widely used as an immunosuppressant in clinic. However, mounting evidence has shown that cyclosporine hinders tolerance induction by dampening Tregs. Therefore, it is of paramount importance to overcome this pitfall. Kaempferol was reported to inhibit DC function. Here, we found that kaempferol delayed islet allograft rejection. Combination of kaempferol and low-dose, but not high-dose, of cyclosporine induced allograft tolerance in majority of recipient mice. Although kaempferol plus either dose of cyclosporine largely abrogated proliferation of graft-infiltrating T cells and their CTL activity, both proliferation and CTL activity in mice treated with kaempferol plus low-dose, but not high-dose, cyclosporine reemerged rapidly upon treatment withdrawal. Kaempferol increased CD4+FoxP3+ Tregs both in transplanted mice and in vitro, likely by suppressing DC maturation and their IL-6 expression. Reduction in Tregs by low dose of cyclosporine was reversed by kaempferol. Kaempferol-induced Tregs exhibited both allospecific and non-allospecific suppression. Administering IL-6 abrogated allograft tolerance induced by kaempferol and cyclosporine via diminishing CD4+FoxP3+ Tregs. Thus, for the first time, we demonstrated that kaempferol promotes transplant tolerance in the presence of low dose of cyclosporine, which allows for sufficient Treg generation while minimizing side effects, resulting in much-needed synergy between kaempferol and cyclosporine.
Collapse
Affiliation(s)
- Y Q Zeng
- Department of Nephrology, the Second Clinical College, Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, P. R. China
| | - X S Liu
- Department of Nephrology, the Second Clinical College, Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, P. R. China
| | - S Wu
- Center for Regenerative and Translational Medicine, the Second Clinical College, Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, P. R. China
| | - C Zou
- Department of Nephrology, the Second Clinical College, Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, P. R. China
| | - Q Xie
- Center for Regenerative and Translational Medicine, the Second Clinical College, Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, P. R. China
| | - S M Xu
- Center for Regenerative and Translational Medicine, the Second Clinical College, Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, P. R. China
| | - X W Jin
- Center for Regenerative and Translational Medicine, the Second Clinical College, Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, P. R. China
| | - W Li
- Center for Regenerative and Translational Medicine, the Second Clinical College, Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, P. R. China
| | - A Zhou
- The Cardiovascular Research Center, Warren Alpert Medical School of Brown University, Providence, RI
| | - Z Dai
- Center for Regenerative and Translational Medicine, the Second Clinical College, Guangzhou University of Chinese Medicine, and Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, Guangdong, P. R. China
| |
Collapse
|
13
|
Payne KK, Bear HD, Manjili MH. Adoptive cellular therapy of cancer: exploring innate and adaptive cellular crosstalk to improve anti-tumor efficacy. Future Oncol 2014; 10:1779-94. [DOI: 10.2217/fon.14.97] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
ABSTRACT The mammalian immune system has evolved to produce multi-tiered responses consisting of both innate and adaptive immune cells collaborating to elicit a functional response to a pathogen or neoplasm. Immune cells possess a shared ancestry, suggestive of a degree of coevolution that has resulted in optimal functionality as an orchestrated and highly collaborative unit. Therefore, the development of therapeutic modalities that harness the immune system should consider the crosstalk between cells of the innate and adaptive immune systems in order to elicit the most effective response. In this review, the authors will discuss the success achieved using adoptive cellular therapy in the treatment of cancer, recent trends that focus on purified T cells, T cells with genetically modified T-cell receptors and T cells modified to express chimeric antigen receptors, as well as the use of unfractionated immune cell reprogramming to achieve optimal cellular crosstalk upon infusion for adoptive cellular therapy.
Collapse
Affiliation(s)
- Kyle K Payne
- Department of Microbiology & Immunology, Virginia Commonwealth University, Massey Cancer Center, Richmond, VA 23298, USA
| | - Harry D Bear
- Department of Microbiology & Immunology, Virginia Commonwealth University, Massey Cancer Center, Richmond, VA 23298, USA
- Department of Surgery, Virginia Commonwealth University, Massey Cancer Center, Richmond, VA 23298, USA
| | - Masoud H Manjili
- Department of Microbiology & Immunology, Virginia Commonwealth University, Massey Cancer Center, Richmond, VA 23298, USA
| |
Collapse
|
14
|
Peripheral blood mononuclear cells of patients with breast cancer can be reprogrammed to enhance anti-HER-2/neu reactivity and overcome myeloid-derived suppressor cells. Breast Cancer Res Treat 2013; 142:45-57. [PMID: 24197563 DOI: 10.1007/s10549-013-2733-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 10/10/2013] [Indexed: 01/04/2023]
Abstract
Two major barriers in the immunotherapy of breast cancer include tumor-induced immune suppression and the establishment of long-lasting immune responses against the tumor. Recently, we demonstrated in an animal model of breast carcinoma that expanding and reprogramming tumor-sensitized lymphocytes, ex vivo, yielded T memory (Tm) cells as well as activated CD25+ NKT cells and NK cells. The presence of activated CD25+ NKT and NK cells rendered reprogrammed T cells resistant to MDSC-mediated suppression, and adoptive cellular therapy (ACT) of reprogrammed lymphocytes protected the host from tumor development and relapse. Here, we performed a pilot study to determine the clinical applicability of our protocol using peripheral blood mononuclear cells (PBMCs) of breast cancer patients, ex vivo. We show that bryostatin 1 and ionomycin combined with IL-2, IL-7, and IL-15 can expand and reprogram tumor-sensitized PBMCs. Reprogrammed lymphocytes contained activated CD25+ NKT and NK cells as well as Tm cells and displayed enhanced reactivity against HER-2/neu in the presence of MDSCs. The presence of activated NKT cells was highly correlated with the rescue of anti-HER-2/neu immune responses from MDSC suppression. Ex vivo blockade experiments suggest that the NKG2D pathway may play an important role in overcoming MDSC suppression. Our results show the feasibility of reprogramming tumor-sensitized immune cells, ex vivo, and provide rationale for ACT of breast cancer patients.
Collapse
|
15
|
Ortiz YM, Arias LF, Álvarez CM, García LF. Memory phenotype and polyfunctional T cells in kidney transplant patients. Transpl Immunol 2013; 28:127-37. [DOI: 10.1016/j.trim.2013.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 02/19/2013] [Accepted: 02/20/2013] [Indexed: 10/27/2022]
|
16
|
Shameli A, Yamanouchi J, Tsai S, Yang Y, Clemente-Casares X, Moore A, Serra P, Santamaria P. IL-2 promotes the function of memory-like autoregulatory CD8+T cells but suppresses their development via FoxP3+Treg cells. Eur J Immunol 2013. [DOI: 10.1002/eji.201242845] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Afshin Shameli
- Julia McFarlane Diabetes Research Centre (JMDRC) and Departments of Microbiology; Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, the University of Calgary; Calgary AB Canada
| | - Jun Yamanouchi
- Julia McFarlane Diabetes Research Centre (JMDRC) and Departments of Microbiology; Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, the University of Calgary; Calgary AB Canada
| | - Sue Tsai
- Julia McFarlane Diabetes Research Centre (JMDRC) and Departments of Microbiology; Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, the University of Calgary; Calgary AB Canada
| | - Yang Yang
- Julia McFarlane Diabetes Research Centre (JMDRC) and Departments of Microbiology; Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, the University of Calgary; Calgary AB Canada
- Department of Biochemistry and Molecular Biology; Faculty of Medicine, the University of Calgary; Calgary AB Canada
| | - Xavier Clemente-Casares
- Julia McFarlane Diabetes Research Centre (JMDRC) and Departments of Microbiology; Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, the University of Calgary; Calgary AB Canada
| | - Anna Moore
- Molecular Imaging Laboratory; MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging; Department of Radiology; Massachusetts General Hospital; Charlestown MA USA
| | - Pau Serra
- Institut d'Investigacions Biomédiques August Pi i Sunyer - Hospital Clinic de Barcelona; Centre Esther Koplowitz Barcelona Spain
| | - Pere Santamaria
- Julia McFarlane Diabetes Research Centre (JMDRC) and Departments of Microbiology; Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, the University of Calgary; Calgary AB Canada
- Institut d'Investigacions Biomédiques August Pi i Sunyer - Hospital Clinic de Barcelona; Centre Esther Koplowitz Barcelona Spain
| |
Collapse
|
17
|
de Chickera S, Willert C, Mallet C, Foley R, Foster P, Dekaban GA. Cellular MRI as a suitable, sensitive non-invasive modality for correlating in vivo migratory efficiencies of different dendritic cell populations with subsequent immunological outcomes. Int Immunol 2011; 24:29-41. [PMID: 22190576 DOI: 10.1093/intimm/dxr095] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The clinical application of dendritic cells (DC) as adjuvants in immunotherapies such as the cell-based cancer vaccine continues to gain interest. The overall efficacy of this emerging immunotherapy, however, remains low. Studies suggest the stage of maturation and activation of ex vivo-prepared DC immediately prior to patient administration is critical to subsequent DC migration in vivo, which ultimately affects overall vaccine efficacy. While it is possible to generate mature and activated DC ex vivo using various stimulatory cocktails, in the case of cancer patients, the qualitative and quantitative assessment of which DC stimulatory cocktail works most effectively to enhance subsequent DC migration in vivo is difficult. Thus, a non-invasive imaging modality capable of monitoring the real-time migration of DC in long-term studies is required. In this paper, we address whether cellular magnetic resonance imaging (MRI) is sufficiently sensitive to quantitatively detect differences in the migratory abilities of two different DC preparations: untreated (resting) versus ex vivo matured in a mouse model. In order to distinguish our ex vivo-generated DC of interest from surrounding tissues in magnetic resonance (MR) images, DC were labeled in vitro with the superparamagnetic iron oxide (SPIO) nanoparticle FeREX®. Characterization of DC phenotype and function following addition of a cytokine maturation cocktail and the toll-like receptor ligand CpG, both in the presence and in the absence of SPIO, were also carried out. Conventional histological techniques were used to verify the quantitative data obtained from MR images. This study provides important information relevant to tracking the in vivo migration of ex vivo-prepared and stimulated DC.
Collapse
Affiliation(s)
- Sonali de Chickera
- BioTherapeutics Research Laboratory, Robarts Research Institute, London, Ontario, N6A 5K8
| | | | | | | | | | | |
Collapse
|
18
|
Lambe T, Crawford G, Johnson AL, Crockford TL, Bouriez-Jones T, Smyth AM, Pham THM, Zhang Q, Freeman AF, Cyster JG, Su HC, Cornall RJ. DOCK8 is essential for T-cell survival and the maintenance of CD8+ T-cell memory. Eur J Immunol 2011; 41:3423-35. [PMID: 21969276 DOI: 10.1002/eji.201141759] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 09/01/2011] [Accepted: 09/23/2011] [Indexed: 01/22/2023]
Abstract
Deficiency in the guanine nucleotide exchange factor dedicator of cytokinesis 8 (DOCK8) causes a human immunodeficiency syndrome associated with recurrent sinopulmonary and viral infections. We have recently identified a DOCK8-deficient mouse strain, carrying an ethylnitrosourea-induced splice-site mutation that shows a failure to mature a humoral immune response due to the loss of germinal centre B cells. In this study, we turned to T-cell immunity to investigate further the human immunodeficiency syndrome and its association with decreased peripheral CD4(+) and CD8(+) T cells. Characterisation of the DOCK8-deficient mouse revealed T-cell lymphopenia, with increased T-cell turnover and decreased survival. Egress of mature CD4(+) thymocytes was reduced with increased migration of these cells to the chemokine CXCL12. However, despite the two-fold reduction in peripheral naïve T cells, the DOCK8-deficient mice generated a normal primary CD8(+) immune response and were able to survive acute influenza virus infection. The limiting effect of DOCK8 was in the normal survival of CD8(+) memory T cells after infection. These findings help to explain why DOCK8-deficient patients are susceptible to recurrent infections and provide new insights into how T-cell memory is sustained.
Collapse
Affiliation(s)
- Teresa Lambe
- Nuffield Department of Medicine, Henry Wellcome Building of Molecular Physiology, Oxford University, Oxford, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Ji H, Shen XD, Gao F, Busuttil RW, Zhai Y, Kupiec-Weglinski JW. Alloreactive CD8 T-cell primed/memory responses and accelerated graft rejection in B-cell-deficient sensitized mice. Transplantation 2011; 91:1075-81. [PMID: 21427633 PMCID: PMC3593653 DOI: 10.1097/tp.0b013e31821578da] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
BACKGROUND The sensitized patients can develop an accelerated form of graft rejection mediated by humoral and T-cell-mediated responses, which are resistant to currently used immunosuppression. METHODS AND RESULTS In our model of fulminant cardiac allograft rejection in sensitized hosts, groups of wild-type (WT) and B-cell-deficient (BKO) mice (B6) were challenged with skin grafts (B/c). Alloreactive CD8 T effector (Teff) activation and T memory (Tmem) differentiation during a 60-day follow-up period were reduced in the absence of B-cell help. The expression of interleukin (IL)-2Rα, IL-7Rα, and IL-15Rα, which support/program CD8 Teff/Tmem expansion, differentiation, and survival, were selectively decreased in BKO hosts. Unlike in WT, in vivo cytotoxic activity analysis of alloreactive Tmem recall response has revealed decreased donor-type (B/c) but not third-party (C3H) cell lysis in sensitized BKO hosts. However, such impaired allo-Ag specific Tmem recall function was insufficient to markedly prolong cardiac allograft survival in sensitized BKO recipients. Indeed, despite quantitative and statistically significant differences between both animal groups, the biological impact of decreased CD8 Teff/Tmem activation and function in the sensitization phase was marginal. Indeed, cardiac allografts underwent fulminant rejection in sensitized BKO, albeit with somewhat delayed kinetics. Interestingly, unlike in naïve counterparts, the rejection cascade remained CD154 blockade-resistant, evidenced by comparable kinetics, and intra-graft cytokine gene profiles in MR1 monoclonal antibody-treated sensitized WT and BKO hosts. CONCLUSION Although B cells were important for optimal alloreactive CD8 Teff/Tmem function in the sensitization phase, the fulminant rejection of cardiac allografts was B-cell-independent, and CD154 blockade-resistant, as in WT hosts.
Collapse
Affiliation(s)
- Haofeng Ji
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Xiu-da Shen
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Feng Gao
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Ronald W. Busuttil
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Yuan Zhai
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| | - Jerzy W. Kupiec-Weglinski
- Dumont-UCLA Transplant Center, Division of Liver and Pancreas Transplantation, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA
| |
Collapse
|
20
|
Goldstein DR. Inflammation and transplantation tolerance. Semin Immunopathol 2011; 33:111-5. [DOI: 10.1007/s00281-011-0251-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2010] [Accepted: 01/13/2011] [Indexed: 12/01/2022]
|
21
|
Zhang S, Dai H, Wan N, Moore Y, Dai Z. Manipulating IL-2 availability amid presentation of donor MHC antigens suppresses murine alloimmune responses by inducing regulatory T cells. PLoS One 2010; 5:e8756. [PMID: 20090908 PMCID: PMC2807454 DOI: 10.1371/journal.pone.0008756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Accepted: 12/18/2009] [Indexed: 12/04/2022] Open
Abstract
Background Major histocompatibility complex (MHC) antigens are important for alloimmune responses as well as immune tolerance. Previous studies have shown that presentation of donor MHC antigens by donor-specific transfusion prior to or upon transplantation promotes transplant tolerance induced by other agents. However, it is unclear whether presentation of donor MHC antigens by DNA vaccination induces long-term allograft survival. Methodology/Principal Findings We investigated whether presentation of MHC class-II and/or class-I donor antigens by DNA vaccination suppresses alloimmune responses and promotes long-term allograft acceptance. We initially found that presentation of both MHC donor antigens by DNA vaccination itself prior to transplantation fails to significantly prolong islet allograft survival in otherwise untreated mice. However, islet allograft survival was significantly prolonged when MHC class-II DNA vaccination was accompanied with IL-2 administration (MHCII + IL-2) while MHC class-I DNA vaccination was followed by IL-2 and subsequent neutralizing anti-IL-2 treatments (MHCI + IL-2/anti-IL-2). Especially, this protocol promoted long-term allograft survival in the majority of recipients (57%) when combined with low doses of rapamycin post-transplantation. Importantly, MHCII + IL-2 induced FoxP3+ Treg cells in both spleens and grafts and suppressed graft-infiltrating CD4+ cell proliferation, whereas MHCI + IL-2/anti-IL-2 mainly inhibited graft-infiltrating CD8+ cell proliferation and donor-specific CTL activity. The combined protocol plus rapamycin treatment further reduced both CD4+ and CD8+ T cell proliferation as well as donor-specific CTL activity but spared FoxP3+ Treg cells. Depleting CD25+ Treg cells or adoptive transfer of pre-sensitized CD8+ T cells abolished this long-term allograft survival. Conclusions/Significance Manipulating IL-2 availability during presentation of MHC class-II and class-I donor antigens by DNA vaccination pre-transplantation induces Treg cells, suppresses alloimmune responses and promotes long-term allograft survival.
Collapse
Affiliation(s)
- Shuzi Zhang
- Center for Biomedical Research, University of Texas Health Science Center, Tyler, Texas, United States of America
| | - Hehua Dai
- Center for Biomedical Research, University of Texas Health Science Center, Tyler, Texas, United States of America
| | - Ni Wan
- Center for Biomedical Research, University of Texas Health Science Center, Tyler, Texas, United States of America
| | - Yolonda Moore
- Center for Biomedical Research, University of Texas Health Science Center, Tyler, Texas, United States of America
| | - Zhenhua Dai
- Center for Biomedical Research, University of Texas Health Science Center, Tyler, Texas, United States of America
- * E-mail:
| |
Collapse
|
22
|
Mays JW, Bailey MT, Hunzeker JT, Powell ND, Papenfuss T, Karlsson EA, Padgett DA, Sheridan JF. Influenza virus-specific immunological memory is enhanced by repeated social defeat. THE JOURNAL OF IMMUNOLOGY 2010; 184:2014-25. [PMID: 20083672 DOI: 10.4049/jimmunol.0900183] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Immunological memory (MEM) development is affected by stress-induced neuroendocrine mediators. Current knowledge about how a behavioral interaction, such as social defeat, alters the development of adaptive immunity, and MEM is incomplete. In this study, the experience of social disruption stress (SDR) prior to a primary influenza viral infection enhanced the frequency and function of the T cell memory pool. Socially stressed mice had a significantly enlarged population of CD8(+) T cells specific for the immunodominant NP366-74 epitope of A/PR/8/34 virus in lung and spleen tissues at 6-12 wk after primary infection (resting memory). Moreover, during resting memory, SDR-MEM mice responded with an enhanced footpad delayed-type hypersensitivity response, and more IFN-gamma-producing CD4(+) T cells were detected after ex vivo stimulation. When mice were rechallenged with A/PR/8/34 virus, SDR-MEM mice terminated viral gene expression significantly earlier than MEM mice and generated a greater D(b)NP(366-74)CD8(+) T cell response in the lung parenchyma and airways. This enhancement was specific to the T cell response. SDR-MEM mice had significantly attenuated anti-influenza IgG titers during resting memory. Similar experiments in which mice were primed with X-31 influenza and challenged with A/PR/8/34 virus elicited similar enhancements in the splenic and lung airway D(b)NP(366-74)CD8(+) T cell populations in SDR-MEM mice. This study demonstrates that the experience of repeated social defeat prior to a primary viral infection significantly enhances virus-specific memory via augmentation of memory T cell populations and suggests that social stressors should be carefully considered in the design and analysis of future studies on antiviral immunity.
Collapse
Affiliation(s)
- Jacqueline W Mays
- Section of Oral Biology, College of Dentistry, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Boyman O, Cho JH, Sprent J. The Role of Interleukin-2 in Memory CD8 Cell Differentiation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 684:28-41. [DOI: 10.1007/978-1-4419-6451-9_3] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
24
|
The role of tryptophan catabolism in acquisition and effector function of memory T cells. Curr Opin Organ Transplant 2008; 13:31-5. [PMID: 18660704 DOI: 10.1097/mot.0b013e3282f3dee1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE OF REVIEW Indoleamine 2,3-dioxygenase (IDO) catalyzes tryptophan and suppresses adaptive immunity by inhibiting T-cell proliferation and promoting their apoptosis. This article reviews the impact of IDO on cellular immunity as well as alloimmunity and particularly discusses the role of tryptophan catabolism in the generation and function of allospecific memory T cells, as the latter pose a long-term threat to allograft survival. RECENT FINDINGS IDO catalyzes tryptophan and suppresses T-cell proliferation while tryptophan metabolites induce T-cell apoptosis. IDO, therefore, suppresses adaptive immunity. Recent studies have shown that IDO overexpression suppresses allograft rejection and autoimmune diseases. Particularly, our new study has shown that IDO is capable of inhibiting the generation and function of allospecific central memory CD8+ T cells, however it does not impair effector memory CD8+ T-cell function. SUMMARY IDO has several immunosuppressive properties that make it a potential candidate for use in transplantation. An effective method to deliver IDO in vivo, however, is lacking. Moreover, IDO alone is insufficient for inducing long-term allograft survival, as high expression of IDO in the graft fails to prevent acute rejection. Further studies are warranted to search for drugs that increase IDO expression in vivo or to explore strategies of prolonging the half life of IDO.
Collapse
|
25
|
Bachmann MF, Oxenius A. Interleukin 2: from immunostimulation to immunoregulation and back again. EMBO Rep 2008; 8:1142-8. [PMID: 18059313 DOI: 10.1038/sj.embor.7401099] [Citation(s) in RCA: 184] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2007] [Accepted: 09/24/2007] [Indexed: 12/18/2022] Open
Abstract
Interleukin 2 (IL-2) was one of the first cytokines to be discovered. However, the complex role of IL-2 and its receptor in the regulation of immune responses is only now emerging. This review explores the various signals triggered by IL-2 and discusses their translation into biological function. A model is outlined that accommodates the seemingly contradictory functions of IL-2, and explains how one cytokine can be an essential T-cell growth and differentiation factor and yet also be indispensable to maintain peripheral tolerance.
Collapse
Affiliation(s)
- Martin F Bachmann
- Cytos Biotechnology AG, Wagistrasse 25, 8952 Zürich-Schlieren, Switzerland.
| | | |
Collapse
|
26
|
Abstract
Much data support an essential role for interleukin (IL)-2 in immune tolerance. This idea is much different from the early paradigm in which IL-2 is central for protective immune responses. This change in thinking occurred when a T regulatory cell defect was shown to be responsible for the lethal autoimmunity associated with IL-2/IL-2R deficiency. This realization allowed investigators to explore immune responses in IL-2-nonresponsive mice rendered autoimmune-free. Such studies established that IL-2 sometimes contributes to optimal primary immune responses, but it is not mandatory. Emerging findings, however, suggest an essential role for IL-2 in immune memory. Here, the current understanding of the dual role of IL-2 in maintaining tolerance and contributing to immunity in vivo is reviewed with some emphasis on T regulatory cell production and homeostasis. Also discussed are implications of this new appreciation concerning the immunobiology of IL-2 with respect to targeting IL-2 or its receptor in immunotherapy.
Collapse
Affiliation(s)
- Thomas R Malek
- Department of Microbiology and Immunology and the Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida 33101, USA.
| |
Collapse
|
27
|
Valujskikh A, Li XC. Frontiers in nephrology: T cell memory as a barrier to transplant tolerance. J Am Soc Nephrol 2007; 18:2252-61. [PMID: 17634436 DOI: 10.1681/asn.2007020151] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Anna Valujskikh
- Department of Immunology, The Cleveland Clinic Foundation, Cleveland, OH 44195, USA.
| | | |
Collapse
|
28
|
Montero E, Alonso L, Perez R, Lage A. Interleukin-2 Mastering Regulation in Cancer and Autoimmunity. Ann N Y Acad Sci 2007; 1107:239-50. [PMID: 17804552 DOI: 10.1196/annals.1381.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Autoimmunity and tumor immunity evolved as two distinct arenas in immunological research. However, the identification of self-antigens as the major components of malignant cells may define a central role for autoimmunity in cancer control tuned by peripheral immunoregulatory mechanisms avoiding self-aggression. Emerging evidence documents a triple antagonistic role of interleukin-2 (IL-2) in vivo promoting survival, apoptosis, and the generation of regulatory T cells. We have found that IL-2 administration reduces the clinical course of experimental autoimmune encephalomyelitis and enhances immunoregulation in tumor-bearing mice. However, actively induced anti-IL-2 antibodies restore the response to nominal antigens in tumor-induced immunosuppressed host and induced therapeutic effect in transplantable and chemically induced tumors. It is suggested that IL-2 may contribute dynamically to the maintenance of natural immunological tolerance, preventing pathological autoimmunity, but may affect antitumor immunity. Cancer research has gained from autoimmunity understanding that tumor escape strategies include the natural mechanisms of immune tolerance and the ways to imbalance the peripheral regulatory mechanisms. Interestingly, therapeutic manipulations of immunoregulation have limited antitumor effects, although promoting collaterally infrequent autoimmune diseases. It may suggest that tumors may reinforce tolerance to protect themselves from the immune attack, a process that may involve dynamically various mechanisms including IL-2. Understanding this acquired experience from tumors may help utilize them to revert the immunopathology in autoimmune diseases.
Collapse
Affiliation(s)
- Enrique Montero
- Center of Molecular Immunology, Experimental Immunotherapy Department, 216 and 15, Playa, P.O. Box: 16040, Havana 11600, Cuba.
| | | | | | | |
Collapse
|
29
|
Abstract
T cell memory induced by prior infection or vaccination provides enhanced protection against subsequent microbial infections. The processes involved in generating and maintaining T cell memory are becoming better understood due to recent technological advances in identifying memory T cells and monitoring their behavior and function in vivo. Memory T cells develop in response to a progressive set of cues-starting with signals from antigen-loaded, activated antigen-presenting cells (APCs) and inflammatory mediators induced by the innate immune response, to the poorly defined subsequent signals triggered as the immune response wanes toward homeostasis. The persistence of the resting memory T cells that eventually develop is regulated by cytokines. This chapter discusses recent findings on how memory T cells develop to confer long-term protective immunity.
Collapse
Affiliation(s)
- J T Tan
- Department of Immunology, The Scripps Research Institute, 10550 North Torrey Pines Rd., La Jolla, CA 92037, USA
| | | |
Collapse
|
30
|
Ndejembi MP, Tang AL, Farber DL. Reshaping the past: Strategies for modulating T-cell memory immune responses. Clin Immunol 2006; 122:1-12. [PMID: 16916619 DOI: 10.1016/j.clim.2006.06.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2006] [Accepted: 06/26/2006] [Indexed: 02/08/2023]
Abstract
Memory T cells are generated following an initial encounter with antigen, persist over the lifetime of an individual, and mediate rapid and robust functional responses upon antigenic recall. While immune memory is generally associated with protective immune response to pathogens, memory T cells can be generated to diverse types of antigens including autoantigens and alloantigens through homologous or crossreactive priming and comprise the majority of circulating T cells during adulthood. Memory T cells can therefore play critical roles in propagating and perpetuating autoimmune disease and in mediating allograft rejection, although the precise pathways for regulation of memory immune responses remain largely undefined. Moreover, evaluating and designing strategies to modulate memory T-cell responses are challenging given the remarkable heterogeneity of memory T cells, with different subsets predominating in lymphoid versus non-lymphoid tissue sites. In this review, we discuss what is presently known regarding the effect of current immunomodulation strategies on the memory T-cell compartment and potential strategies for controlling immunological recall.
Collapse
Affiliation(s)
- Modesta P Ndejembi
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | |
Collapse
|
31
|
Kamimura D, Sawa Y, Sato M, Agung E, Hirano T, Murakami M. IL-2 in vivo activities and antitumor efficacy enhanced by an anti-IL-2 mAb. THE JOURNAL OF IMMUNOLOGY 2006; 177:306-14. [PMID: 16785526 DOI: 10.4049/jimmunol.177.1.306] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
IL-2 is a potent immunostimulant and has been tested for clinical use, including in immunotherapy for cancers and HIV infection. Here we show that a widely used neutralizing anti-murine IL-2 mAb (S4B6) exhibits unexpected activities that enhance the treatment effects of IL-2 in vivo. Coinjection of the anti-IL-2 mAb with a plasmid carrying murine IL-2 cDNA significantly increased the serum IL-2 levels and induced a substantial increase in the division of CD8+ T and NK1.1(high) cells in vivo. Injection of the mAb premixed with recombinant murine IL-2 showed the same enhanced effect. A 5-day treatment with the anti-IL-2 mAb alone gradually increased the CD44(high)CD8+ population, and the increased population was maintained for >300 days, suggesting that the mAb can gradually maintain and potentially enhance the bioactivity of endogenous IL-2 for extended periods. Furthermore, combined treatment with the anti-IL-2 mAb plus the IL-2 plasmid markedly enhanced Ag-specific CTL activity in vivo and partially protected mice from tumor metastasis to the lungs, compared with the anti-IL-2 mAb or IL-2 plasmid alone. These results demonstrated IL-2-enhancing effects of the anti-IL-2 mAb in vivo and suggest that combining a neutralizing anti-IL-2 Ab with IL-2 gene delivery might be used effectively to enhance IL-2 functions in clinical applications.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/physiology
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/physiology
- Antigens, Ly
- Antigens, Surface/biosynthesis
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/agonists
- Antineoplastic Agents/blood
- Antineoplastic Agents/immunology
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Line, Tumor
- Down-Regulation/genetics
- Down-Regulation/immunology
- Forkhead Transcription Factors/antagonists & inhibitors
- Forkhead Transcription Factors/biosynthesis
- Hyaluronan Receptors/biosynthesis
- Interleukin-2/administration & dosage
- Interleukin-2/blood
- Interleukin-2/genetics
- Interleukin-2/immunology
- Killer Cells, Natural/cytology
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Lectins, C-Type/biosynthesis
- Lung Neoplasms/immunology
- Lung Neoplasms/secondary
- Lung Neoplasms/therapy
- Lymphocyte Count
- Melanoma, Experimental/immunology
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- NK Cell Lectin-Like Receptor Subfamily B
- Neoplasm Transplantation
- Recombinant Proteins/administration & dosage
- Recombinant Proteins/agonists
- Recombinant Proteins/blood
- Recombinant Proteins/immunology
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
Collapse
Affiliation(s)
- Daisuke Kamimura
- Laboratory for Cytokine Signaling, RIKEN Research Center for Allergy and Immunology, Yokohama, Kanagawa, Japan
| | | | | | | | | | | |
Collapse
|
32
|
Spierings DCJ, Lemmens EE, Grewal K, Schoenberger SP, Green DR. Duration of CTL activation regulates IL-2 production required for autonomous clonal expansion. Eur J Immunol 2006; 36:1707-17. [PMID: 16791878 DOI: 10.1002/eji.200635929] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The extent and magnitude of CD8(+) T lymphocyte (CTL) clonal expansion in vivo depends on the duration of stimulation provided during primary activation, whereas expansion under conventional in vitro conditions fails to reveal this. The molecular details of this differential programming are unclear. We developed a low-density culture system that recapitulates in vivo conditions and enables analysis of both proliferation and survival/apoptosis during primary expansion. We found that CTL given a prolonged stimulus produced more IL-2 and for a longer period than briefly activated CTL, and the IL-2 produced during the expansion phase was essential for cell proliferation and accumulation at low densities. CTL given a short stimulation and plated at low density followed the fate of briefly stimulated cells in vivo and underwent an abortive expansion and apoptosis, unless IL-2 was exogenously provided. Inhibition of apoptosis did not replace the requirement for IL-2 to drive clonal expansion. CTL primed for prolonged periods but in the absence of costimulation similarly behaved as though "partially programmed", undergoing abortive expansion at low densities. These results indicate that the nature and duration of antigenic stimulation regulate the autonomy of CTL expansion via the production of autocrine IL-2.
Collapse
|
33
|
Lucas PJ, Kim SJ, Mackall CL, Telford WG, Chu YW, Hakim FT, Gress RE. Dysregulation of IL-15-mediated T-cell homeostasis in TGF-beta dominant-negative receptor transgenic mice. Blood 2006; 108:2789-95. [PMID: 16788095 PMCID: PMC1895588 DOI: 10.1182/blood-2006-05-025676] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
T-cell subpopulations, defined by their expression of CD4, CD8, naive, and memory cell-surface markers, occupy distinct homeostatic compartments that are regulated primarily by cytokines. CD8+ memory T cells, as defined by CD44(hi) surface expression, are dependent on IL-15 as a positive regulator of their homeostatic maintenance. Manipulation of IL-15 signaling through gene aberration, overexpression, or receptor alterations has been shown to dramatically affect T-cell homeostasis, with overexpression leading to fatal leukemia. Here we show that TGF-beta is the critical negative regulator of murine CD8+ memory T-cell homeostasis with direct opposition to the positive effects of IL-15. This negative regulation is mediated, at least in part, by the ability of TGF-beta to modulate expression of the beta-chain of the IL-15 receptor, thus establishing a central axis between these 2 cytokines for homeostatic control of CD8+ memory T-cell populations. These data establish TGF-beta as a critical and dominant tumor-suppressor pathway opposing IL-15-mediated CD8+ T-cell expansion and potential malignant transformation.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Female
- Genes, T-Cell Receptor
- Homeostasis
- Immunologic Memory
- Interleukin-15/deficiency
- Interleukin-15/genetics
- Interleukin-15/metabolism
- Interleukin-2/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Protein Serine-Threonine Kinases
- Receptor, Transforming Growth Factor-beta Type II
- Receptors, Interleukin-15
- Receptors, Interleukin-2/chemistry
- Receptors, Interleukin-2/genetics
- Receptors, Interleukin-2/metabolism
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/metabolism
- Signal Transduction
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
Collapse
Affiliation(s)
- Philip J Lucas
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, CRC/3-3288, 10 Center Drive, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
During T-cell priming, cytokines and costimulatory molecules provide important signals that determine the magnitude and quality of the response. Although the functions of defined cytokines and costimulators in the primary T-cell response are well characterized, much less is known about how these factors contribute to memory T-cell development and survival. Since memory cells are thought to be long-lived progeny of the primary response, it is conceivable that the same signals shaping initial T-cell expansion and differentiation also contribute to memory generation. Here, we review evidence and show novel data on the role of the cytokines interleukin-2 (IL-2) and IL-7 and the costimulator CD28 in CD4+ memory T-cell development. We emphasize that transient IL-2 and CD28 signals during priming imprint a long-lasting survival advantage in primed T cells, thus contributing to the persistence of a memory population. The requirement for IL-2 and CD28 signals is not linked to promoting T-cell division and expansion but most likely due to their capacity to (i) promote effector cell differentiation; (ii) induce survival proteins, and, as we discuss in more detail; (iii) program expression of receptors for 'memory survival factors' such as IL-7. Studies exploring the therapeutic potential of these insights are also discussed.
Collapse
Affiliation(s)
- Hans Dooms
- Department of Pathology, University of California-San Francisco, San Francisco, CA 94143-0511, USA
| | | |
Collapse
|
35
|
Ferrari V, Cacere CR, Machado CM, Pannuti CS, Dulley FL, Barros JCA, Duarte AJDS, Benard G. Distinct patterns of regeneration of central memory, effector memory and effector TCD8+ cell subsets after different hematopoietic cell transplant types: Possible influence in the recovery of anti-cytomegalovirus immune response and risk for its reactivation. Clin Immunol 2006; 119:261-71. [PMID: 16426894 DOI: 10.1016/j.clim.2005.12.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2005] [Revised: 11/29/2005] [Accepted: 12/01/2005] [Indexed: 11/20/2022]
Abstract
TCD8+ cells may be divided into subsets with different phenotypes and functions: naive, central memory, effector memory and effector. Aiming to better understand the differences in early reconstitution of these TCD8+ cell subsets and their relationship with post-transplant anti-cytomegalovirus (CMV) immune responses recovery, we prospectively analyzed the transfer and expansion of these subsets in different transplant types. We found that graft cells from donor's peripheral blood, either allogeneic or autologous, were enriched for central memory, effector memory and effector phenotypes compared to allogeneic bone marrow grafts, as assessed by surface markers phenotyping and granzyme B expression. However, post-transplant, these subsets expanded in autologous recipients only, reaching numbers much greater than in allo-recipients at days +29 and +96. At the same time, autologous recipients presented less CMV reactivation and more vigorous CMV-induced interferon-gamma and lymphoproliferative responses. The marked loss of allo-transferred memory TCD8+ cells was probably due to the fact that they were more activated and more prone to apoptosis than auto-transferred TCD8+ cells as assessed by CD69 and active caspase 3 expression. Thus, transfer of peripheral blood stem cells in the allogeneic but not autologous setting is associated with poor expansion of memory TCD8+ cells, probably delaying antiviral immune reconstitution. These data may have important implications for the design of better strategies to immunoprotect this population against infectious challenges since different transplant types have different potentials for memory TCD8+ cells transfer and expansion.
Collapse
Affiliation(s)
- Valeria Ferrari
- Laboratory of Dermatology and Immunodeficiencies (LIM-56), Medical School of the University of São Paulo, Av. Dr. Enéas de Carvalho Aguiar 500 3rd Floor, São Paulo, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Ciupe MS, Bivort BL, Bortz DM, Nelson PW. Estimating kinetic parameters from HIV primary infection data through the eyes of three different mathematical models. Math Biosci 2006; 200:1-27. [PMID: 16469337 DOI: 10.1016/j.mbs.2005.12.006] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2003] [Revised: 12/12/2005] [Accepted: 12/18/2005] [Indexed: 10/25/2022]
Abstract
The dynamics of HIV-1 infection consist of three distinct phases starting with primary infection, then latency and finally AIDS or drug therapy. In this paper we model the dynamics of primary infection and the beginning of latency. We show that allowing for time delays in the model better predicts viral load data when compared to models with no time delays. We also find that our model of primary infection predicts the turnover rates for productively infected T cells and viral totals to be much longer than compared to data from patients receiving anti-viral drug therapy. Hence the dynamics of the infection can change dramatically from one stage to the next. However, we also show that with the data available the results are highly sensitive to the chosen model. We compare the results using analysis and Monte Carlo techniques for three different models and show how each predicts rather dramatic differences between the fitted parameters. We show, using a chi(2) test, that these differences between models are statistically significant and using a jackknifing method, we find the confidence intervals for the parameters. These differences in parameter estimations lead to widely varying conclusions about HIV pathogenesis. For instance, we find in our model with time delays the existence of a Hopf bifurcation that leads to sustained oscillations and that these oscillations could simulate the rapid turnover between viral strains and the appropriate CTL response necessary to control the virus, similar to that of a predator-prey type system.
Collapse
Affiliation(s)
- M S Ciupe
- Los Alamos National Lab, MS K710, Los Alamos, NM 87545, United States
| | - B L Bivort
- Department of Molecular and Cellular Biology, Harvard University, 7 Divinity Avenue, Cambridge, MA 02138, United States
| | - D M Bortz
- Math Biology Research Group, Department of Mathematics, University of Michigan, 530 Church Street, Ann Arbor, MI 48109, United States
| | - P W Nelson
- Los Alamos National Lab, MS K710, Los Alamos, NM 87545, United States; Math Biology Research Group, Department of Mathematics, University of Michigan, 530 Church Street, Ann Arbor, MI 48109, United States.
| |
Collapse
|
37
|
Parameswaran N, Suresh R, Bal V, Rath S, George A. Lack of ICAM-1 on APCs during T cell priming leads to poor generation of central memory cells. THE JOURNAL OF IMMUNOLOGY 2005; 175:2201-11. [PMID: 16081787 DOI: 10.4049/jimmunol.175.4.2201] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
ICAM-1/LFA-1 interactions are known to enhance T cell/APC interactions and to promote T cell activation and cytokine secretion. We have analyzed the consequences of ICAM-1-mediated signaling on the generation of memory T cell subsets. We report that lack of ICAM-1 on APCs, but not on T cells, leads to poor T cell activation and proliferation in vitro and in vivo, and that the defect can be compensated by Ag dose, exogenous IL-2, additional costimulation, and by increasing responder T cell density on APCs. ICAM-1-null mice do not respond to immunization with OVA peptide, but immunization with OVA or with Salmonella typhimurium leads to good T cell proliferation 7-10 days later, and clearance of a challenge infection is equivalent to that of wild-type mice. However, when followed over time, recall proliferation and antibacterial immunity decay rapidly in ICAM-1-null mice, while recall cytokine responses are unaffected. The decline in immunity is not related to poor survival of T cells activated on ICAM-1-null APCs, or to poor generation of effectors in ICAM-1-null mice. Phenotypic analysis of T cells stimulated on ICAM-1-null APCs reveals preferential generation of CD44(high) CD62L(low) effector memory cells (T(EM)) over CD44(high) CD62L(high) central memory cells (T(CM)). Further, while the proportion of naive:memory T cells is similar in unmanipulated wild-type and ICAM-1-null mice, there is an accumulation of T(EM) cells, and a high T(EM):T(CM) ratio in aging ICAM-1-null mice. Together, the data indicate that signaling through LFA-1 during T cell activation may be involved in commitment to a proliferation-competent memory pool.
Collapse
|
38
|
Cleven HA, Genden EM, Moran TM. Reepithelialized orthotopic tracheal allografts expand memory cytotoxic T lymphocytes but show no evidence of chronic rejection. Transplantation 2005; 79:861-8. [PMID: 15849536 DOI: 10.1097/01.tp.0000157119.39395.c3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Acute rejection of mouse tracheal allografts is characterized by infiltration of the lamina propria with CD4+/CD8+ T cells that leads to the destruction of the epithelium and luminal obliteration. The donor epithelium is progressively replaced by recipient-derived epithelium. Once allograft reepithelialization has occurred, immunosuppression can be withdrawn without inciting acute rejection. We hypothesize that reepithelialization will also prevent chronic rejection of the trachea after withdrawal of immunosuppression. METHODS BALB/c tracheal grafts were transplanted orthotopically into allogeneic C57BL/6 recipients. Allografted mice were nonimmunosuppressed for 10 or 100 days or immunosuppressed with cyclosporine A continuously for 50 days and then withdrawn from immunosuppression for an additional 50 days. In addition, grafts from this group were then heterotopically retransplanted into isogenic C57BL/6 or allogeneic BALB/c recipients to assess their immunogenicity. RESULTS Cyclosporine A-treated mice showed no signs of chronic rejection or priming of cellular immunity as measured by proliferation and cytokine secretion in a mixed leukocyte reaction. However, there was a notable expansion of memory CD8+ T cells specific for donor major histocompatibility complex. When these tracheal allografts were retransplanted heterotopically into C57BL/6 or BALB/c, they demonstrated reduced responses toward BALB/c and primed responses toward C57BL/6, respectively. These results suggest that the grafts express a chimeric phenotype consisting of both BALB/c and C57BL/6 antigens. CONCLUSION These observations suggest that long-term withdrawal of immunosuppression does not lead to chronic tracheal rejection even in the presence of alloantigen specific cytotoxic T-lymphocyte responses and that the reepithelialized grafts may contain donor elements that impact the generation of immunity.
Collapse
Affiliation(s)
- Heidi A Cleven
- Mount Sinai School of Medicine, Department of Microbiology, New York, New York 10029, USA
| | | | | |
Collapse
|
39
|
Taylor DK, Neujahr D, Turka LA. Heterologous immunity and homeostatic proliferation as barriers to tolerance. Curr Opin Immunol 2005; 16:558-64. [PMID: 15341999 DOI: 10.1016/j.coi.2004.07.007] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The different threshold of activation for memory T cells compared to that of naïve T cells makes them resistant to immunomodulation, thus representing a barrier to tolerance. Recently it has been demonstrated that homeostatic proliferation and heterologous immunity represent two naturally occurring and distinct processes that can generate memory T cells. Homeostatic proliferation refers to the process by which, in a lymphodeficient host, normal T cells 'spontaneously' proliferate in response to self-MHC-peptide complexes. Heterologous immunity refers to a process in which a response to one or more infectious agents generates effector/memory T cells with cross-reactive specificities. Recent new studies have defined the importance of these processes in transplantation models and implicated strategies to induce transplantation tolerance.
Collapse
Affiliation(s)
- Devon K Taylor
- University of Pennsylvania, 700 Clinical Research Building, 415 Curie Boulevard, Philadelphia 19104-6144, USA
| | | | | |
Collapse
|
40
|
Villinger F, Miller R, Mori K, Mayne AE, Bostik P, Sundstrom JB, Sugimoto C, Ansari AA. IL-15 is superior to IL-2 in the generation of long-lived antigen specific memory CD4 and CD8 T cells in rhesus macaques. Vaccine 2004; 22:3510-21. [PMID: 15308379 DOI: 10.1016/j.vaccine.2003.07.022] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2003] [Revised: 07/26/2003] [Accepted: 07/28/2003] [Indexed: 11/17/2022]
Abstract
Using tetanus toxoid (TT) and influenza (Flu) immunization of rhesus macaques as a model, the effect of IL-2 and IL-15 on the generation and maintenance of antigen specific memory T cells was evaluated following primary and secondary immunization. Daily cytokine administration expanded primarily effector but not memory cells, while spacing cytokine administration to q3-7 days markedly enhanced TT and Flu specific memory responses. Following primary immunization, TT specific CD4 and influenza matrix protein (Flu-MP) specific CD8 effector responses were enhanced by IL-2 administration but CD8 specific memory responses were no different from cytokine non-treated monkeys. In contrast, expansion of Flu specific CD8 cells with IL-15 was only modest but resulted in significantly elevated levels of memory cells at 6 months. IL-15 also significantly enhanced early and late TT specific CD4 responses. The highest levels of primary effector and memory T cells were observed following alternate administration of both IL-2 and IL-15. Following booster immunization, however, only IL-15 appeared able to enhance CD8 T cell responses while IL-2 or IL2/IL-15 administration were less effective.
Collapse
Affiliation(s)
- F Villinger
- Department of Pathology and Laboratory Medicine, Emory University, Woodruff Memorial Building 2339, 1639 Pierce Drive, Atlanta, GA 30322, USA.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Sojka DK, Bruniquel D, Schwartz RH, Singh NJ. IL-2 secretion by CD4+ T cells in vivo is rapid, transient, and influenced by TCR-specific competition. THE JOURNAL OF IMMUNOLOGY 2004; 172:6136-43. [PMID: 15128800 DOI: 10.4049/jimmunol.172.10.6136] [Citation(s) in RCA: 123] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The secretion of IL-2 is a critical and early landmark in the activation program of CD4(+) T cells in vitro, but the lack of sensitive assays has limited its application for studying T cell activation in vivo. Using a mouse cytokine capture assay we were able to detect the rapid secretion of IL-2 after an in vivo stimulus by 1-2 h in naive T cells and as early as 30 min in memory T cells. Maximal secretion was achieved within 1-2 h for memory cells or 6-8 h for naive T cells. Surprisingly IL-2 production terminated quickly in vivo and secretion was undetectable by 20-24 h in either cell type. We further demonstrated that this short duration of secretion can be influenced by cellular competition between Ag-specific CD4(+) T cells. The consequences of competition were mimicked by reducing the strength of the antigenic stimulus. These data argue that early competition between T cells influences both the eventual frequency of IL-2 producers in the population and also the duration of their secretion, potentially by altering the strength or duration of the stimulus available to each T cell.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigens, Bacterial/administration & dosage
- Antigens, Bacterial/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cell Communication/genetics
- Cell Communication/immunology
- Clone Cells
- Dose-Response Relationship, Immunologic
- Enterotoxins/administration & dosage
- Enterotoxins/immunology
- Epitopes, T-Lymphocyte/immunology
- Injections, Intraperitoneal
- Interleukin-2/biosynthesis
- Interleukin-2/metabolism
- Kinetics
- Lymphocyte Activation
- Lymphocyte Count
- Lymphocyte Depletion
- Mice
- Mice, Inbred A
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Staphylococcus aureus/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/transplantation
Collapse
Affiliation(s)
- Dorothy K Sojka
- Laboratory of Cellular and Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
42
|
Shinoda K, Xin KQ, Jounai N, Kojima Y, Tamura Y, Okada E, Kawamoto S, Okuda K, Klinman D, Okuda K. Polygene DNA vaccine induces a high level of protective effect against HIV-vaccinia virus challenge in mice. Vaccine 2004; 22:3676-90. [PMID: 15315847 DOI: 10.1016/j.vaccine.2004.03.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2003] [Accepted: 03/14/2004] [Indexed: 11/21/2022]
Abstract
Single HIV-1 subtype DNA vaccine is unlikely to provide reactive protection across a wide range of HIV strains since the HIV virus changes the antigenic sites, particularly, in env gene. To overcome these issues, we constructed a multivalent poly-epitope DNA vaccine. A polygenic DNA vaccine encoding 20 antigenic epitopes from the HIV-1 Env, Gag, and Pol proteins of several clades was constructed using humanized and optimized codons and it was named here hDNA vaccine. In mice, this hDNA vaccine stimulated the following strong (1) antigen-specific serum antibody (Ab) responses, (2) delayed-type hypersensitivity, (3) the activation of IFN-gamma secretion cells targeting gp120 and synthetic antigenic peptides, in addition (4) a significant level of several peptide specific cytotoxic T lymphocytes (CTL) responses. Challenged with modified vaccinia viruses vPE16 and vP1206 expressing HIV-1 env and gag.pol genes, respectively, demonstrated the viral titers in the ovary of the mice vaccinated with hDNA significantly less compared to the unvaccinated mice. Thus, the use of polygene DNA vaccine appears to induce a high level of HIV-specific immune responses and is very effective against challenge with recombinant HIV-vaccinia viruses.
Collapse
MESH Headings
- AIDS Vaccines/immunology
- AIDS Vaccines/therapeutic use
- Amino Acid Sequence
- Animals
- Antibody Formation/immunology
- Cytokines/metabolism
- Enzyme-Linked Immunosorbent Assay
- Gene Products, gag/immunology
- HIV Antibodies/analysis
- HIV Antibodies/biosynthesis
- HIV Envelope Protein gp120/immunology
- HIV Infections/prevention & control
- HIV-1/genetics
- HIV-1/immunology
- Hypersensitivity, Delayed/immunology
- Image Processing, Computer-Assisted
- Immunity, Cellular/immunology
- Immunization
- Interferon-gamma/immunology
- Mice
- Mice, Inbred BALB C
- Molecular Sequence Data
- Plasmids/genetics
- Plasmids/immunology
- Promoter Regions, Genetic/genetics
- T-Lymphocytes, Cytotoxic/immunology
- Vaccines, DNA/immunology
- Vaccines, Subunit/genetics
- Vaccines, Subunit/immunology
- Vaccinia virus/immunology
Collapse
Affiliation(s)
- Kaori Shinoda
- Department of Bacteriology, Yokohama City University School of Medicine, 3-9 Fukuura, Kanazawa-ku, Yokohama 236-0004, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Tesar BM, Zhang J, Li Q, Goldstein DR. TH1 immune responses to fully MHC mismatched allografts are diminished in the absence of MyD88, a toll-like receptor signal adaptor protein. Am J Transplant 2004; 4:1429-39. [PMID: 15307830 DOI: 10.1111/j.1600-6143.2004.00544.x] [Citation(s) in RCA: 108] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Toll-like receptors (TLRs) are innate immune receptors that are critical for recognizing conserved microbial motifs by inducing TH1 immunity. The majority of TLRs utilize the adaptor protein MyD88 for signal transduction, although other adaptors have been recently described. As the role of innate immunity in transplantation is unclear, we examined the importance of the MyD88 pathway in acute rejection of fully MHC-mismatched murine allografts and specifically investigated whether MyD88 signaling is important for DC (dendritic cell) function and TH1 alloimmune responses. Our results demonstrate that acute rejection of both fully allogeneic skin and cardiac allografts occurs in the absence of MyD88. However, priming of naïve recipient T cells by allogeneic DCs and TH1 immune responses were diminished in the absence of MyD88, although TH2 immunity remained intact. Thus, these results demonstrate that MyD88 signaling is important for DC function and TH1 responses during fully MHC-mismatched solid-organ transplantation, although graft rejection occurs independently of MyD88.
Collapse
Affiliation(s)
- Bethany M Tesar
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, USA
| | | | | | | |
Collapse
|
44
|
Teague RM, Tempero RM, Thomas S, Murali-Krishna K, Nelson BH. Proliferation and Differentiation of CD8+ T Cells in the Absence of IL-2/15 Receptor β-Chain Expression or STAT5 Activation. THE JOURNAL OF IMMUNOLOGY 2004; 173:3131-9. [PMID: 15322173 DOI: 10.4049/jimmunol.173.5.3131] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Major gains in the efficacy of T cell-based therapies for cancer and infectious diseases could be realized through improved understanding of the signals that control expansion and differentiation of CD8(+) cytolytic T cells. IL-2, IL-15, and the downstream transcription factor STAT5 have all been implicated as important regulators of these processes, yet there are conflicting data regarding their contribution to in vivo T cell responses. We used a murine adoptive T cell transfer model to examine the contribution of IL-2 and IL-15 signaling to the proliferation and differentiation of naive, CD8(+) T cells bearing an OVA-specific TCR transgene (OT-I). OT-I T cells failed to express the high affinity IL-2R (CD25) while proliferating in vivo, irrespective of the mode of Ag delivery. Moreover, OT-I T cells rendered genetically deficient in the shared IL-2/IL-15Rbeta subunit (IL-2Rbeta) demonstrated normal Ag-induced proliferation and cytolytic activity in vivo. Accordingly, activation of STAT5 was not detected in proliferating IL-2Rbeta-deficient OT-I T cells, thus implicating a STAT5-independent cytokine or costimulatory pathway in this process. Even though IL-2 and IL-15 were dispensable for CD8(+) T cell proliferation, systemic infusion of IL-2 nevertheless promoted the expansion of OT-I T cells in vivo. Thus, IL-2 and IL-15 signals are not essential for CD8(+) T cell proliferation or differentiation, but IL-2 can promote supraphysiological expansion when supplied exogenously. These findings challenge current models that place CD8(+) T cell proliferation under the control of STAT5-dependent cytokines and suggest new approaches to the therapeutic manipulation of T cell numbers in vivo.
Collapse
Affiliation(s)
- Ryan M Teague
- Benaroya Research Institute, Virginia Mason, Seattle, WA, USA
| | | | | | | | | |
Collapse
|
45
|
Kan-Mitchell J, Bisikirska B, Wong-Staal F, Schaubert KL, Bajcz M, Bereta M. The HIV-1 HLA-A2-SLYNTVATL is a help-independent CTL epitope. THE JOURNAL OF IMMUNOLOGY 2004; 172:5249-61. [PMID: 15100263 DOI: 10.4049/jimmunol.172.9.5249] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The CTL response to the HLA-A*0201-restricted, HIV-1 p17 Gag(77-85) epitope (SLYNTVATL; SL9) has been extensively studied in patients. Although this reactivity is exceptionally prominent in chronically infected patients and inversely correlated to viral load, SL9-specific CTLs (SL9-CTLs) are rarely detected in acute infection. To explore the cellular basis for this unusual manifestation, SL9-CTLs primed ex vivo from naive circulating CD8(+) T cells of healthy, seronegative donors were generated and characterized. SL9 appeared to differ from other well-studied A*0201-restricted epitopes in several significant respects. In contrast to published reports for influenza and melanoma peptides and the HIV gag IV9 epitope studied here in parallel, SL9-CTLs were primed by immature but not mature autologous dendritic cells. Highly activated SL9-CTLs produce sufficient autocrine mediators to sustain clonal expansion and CTL differentiation for months without CD4(+) T cells or exogenous IL-2. Moreover, SL9-CTLs were sensitive to paracrine IL-2-induced apoptosis. IL-2 independence and sensitivity to paracrine IL-2 were also characteristic of SL9-CTLs immunized by dendritic cells transduced by a nonreplicating lentiviral vector encoding full-length Gag. In vitro-primed SL9-CTLs resembled those derived from patients in degeneracy of recognition and functional avidities for both SL9 and its natural mutations. Together, these data show that SL9 is a highly immunogenic, help-independent HIV epitope. The scarcity of SL9-CTLs in acute infection may result from cytokine-induced apoptosis with the intense activation of the innate immunity. In contrast, SL9-CTLs that constitutively produce autocrine help would predominate during CD4-diminished chronic infection.
Collapse
MESH Headings
- Antigen Presentation/genetics
- Antigen Presentation/immunology
- Cell Differentiation/immunology
- Cell Division/immunology
- Clone Cells
- Cytokines/physiology
- Cytotoxicity Tests, Immunologic
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Epitopes, T-Lymphocyte/immunology
- Gene Products, gag/immunology
- Genes, T-Cell Receptor beta
- Genetic Vectors
- HIV Antigens/immunology
- HIV-1/genetics
- HIV-1/immunology
- HLA-A Antigens/immunology
- HLA-A2 Antigen/immunology
- Humans
- Interleukin-2/pharmacology
- Lymphocyte Activation/immunology
- Lymphocyte Depletion
- Mutation
- Paracrine Communication/immunology
- Peptide Fragments
- Peptides/immunology
- Peptides/pharmacology
- Receptors, Antigen, T-Cell, alpha-beta/biosynthesis
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Cytotoxic/virology
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/metabolism
- Transduction, Genetic
- Viral Proteins/immunology
- env Gene Products, Human Immunodeficiency Virus
- gag Gene Products, Human Immunodeficiency Virus
Collapse
Affiliation(s)
- June Kan-Mitchell
- Karmanos Cancer Institute, Department of Pathology and Immunology, Wayne State University, Detroit, MI 48201, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Abstract
A cardinal feature of the adaptive immune response is its ability to generate long-lived populations of memory T lymphocytes. Memory T cells are specific to the antigen encountered during the primary immune response and react rapidly and vigorously upon re-encounter with the same antigen. Memory T cells that recognize microbial antigens provide the organism with long-lasting protection against potentially fatal infections. On the other hand, memory T cells that recognize donor alloantigens can jeopardize the survival of life-saving organ transplants. We review here the immunobiology of memory T cells and describe their role in the rejection of solid organ allografts.
Collapse
Affiliation(s)
- Anna Valujskikh
- Department of Immunology, The Cleveland Clinic Foundation, 9500 Euclid Avenue NB30, Cleveland, OH 44195, USA.
| | | |
Collapse
|
47
|
Bingaman AW, Farber DL. Memory T cells in transplantation: generation, function, and potential role in rejection. Am J Transplant 2004; 4:846-52. [PMID: 15147417 DOI: 10.1111/j.1600-6143.2004.00453.x] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The adaptive immune system is endowed with long-lived memory to recall previous antigen encounters and respond more effectively to them. Memory immune responses are mediated by antigen-specific memory T lymphocytes that exhibit enhanced function compared with naïve T cells that have never encountered antigen. While the generation of memory T cells specific for pathogens is beneficial in providing protective immunity, memory T cells specific for alloantigens can be deleterious to the recipient of a transplanted organ. In graft rejection, memory T cells mediate accelerated, "second-set" rejection and their presence has been associated with increased propensity for early rejection. Recent findings have demonstrated that alloreactive memory T cells can be generated via exposure to alloantigens, as well as stimuli that are cross-reactive with alloantigens, and are therefore likely present in "naïve" individuals. This review focuses on the characteristics of memory T cells which make them of special interest to the transplant community, including differential activation requirements, broad homing properties, and resistance to tolerance induction. The multiple ways in which memory T cells can contribute to early and late graft rejection are discussed, as well as potential targets for combating alloreactive memory to be considered in the future design of tolerance induction strategies.
Collapse
Affiliation(s)
- Adam W Bingaman
- Department of Surgery, Division of Transplantation, University of Maryland School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
48
|
Lustgarten J, Dominguez AL, Cuadros C. The CD8+
T cell repertoire against Her-2/neu antigens in neu transgenic mice is of low avidity with antitumor activity. Eur J Immunol 2004; 34:752-761. [PMID: 14991605 DOI: 10.1002/eji.200324427] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The majority of tumor-associated antigens are aberrantly expressed or overexpressed normal gene products. Therefore, mechanisms responsible for self tolerance dampen immune responses against these antigens. To evaluate the effect that tolerance has on the immune responses against tumor antigens, we characterized the CD8+ T cell responses in neu mice. T cell responses against the A2.1/neu p369-377 and p773-782 peptides were evaluated in neu mice that were crossed with A2.1/Kb transgenic mice (A2 x neu). Tetramer binding and cytotoxic activity demonstrate that, compared to CTL from A2.1/Kb x FVB wild-type mice (A2 x FVB), CD8+ T cells from A2 x neu mice were of lower avidity for the peptides. Despite the fact that A2 x neu mice are tolerant, multiple immunizations with DC pulsed with the p369-377 or p773-782 peptides in the presence of IL-2 retarded tumor growth in A2 x neu mice, and immunizations in combination with the anti-OX40 mAb further enhanced the antitumor response. Taken together, these data indicate that low-avidity T cells for neu antigens persisting in A2 x neu mice have the capacity to develop antitumor responses as long as they are provided with efficient costimulation. These results underscore the potential role of low-avidity T cells in antitumor immunity and may offer an important component for vaccination immunotherapies.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antibodies, Monoclonal/pharmacology
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Cell Division
- Cell Line, Tumor
- Dendritic Cells/transplantation
- HLA-A2 Antigen/genetics
- Immune Tolerance
- Mice
- Mice, Transgenic
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Peptides/immunology
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Receptors, OX40
- Receptors, Tumor Necrosis Factor/antagonists & inhibitors
- Receptors, Tumor Necrosis Factor/immunology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/transplantation
Collapse
|
49
|
Chang J, Cho JH, Lee SW, Choi SY, Ha SJ, Sung YC. IL-12 Priming during In Vitro Antigenic Stimulation Changes Properties of CD8 T Cells and Increases Generation of Effector and Memory Cells. THE JOURNAL OF IMMUNOLOGY 2004; 172:2818-26. [PMID: 14978082 DOI: 10.4049/jimmunol.172.5.2818] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Antigenic and costimulatory signals trigger a developmental program by which naive CD8 T cells differentiate into effector and memory cells. However, initial cytokine signals that regulate the generation of effector and memory CD8 T cells are not well understood. In this study, we show that IL-12 priming during in vitro antigenic stimulation results in the significant increase of both primary and memory CD8 T cell population in mice after adoptive transfer of activated cells. The effect of IL-12 priming is closely associated with qualitative changes in CD8 T cells, such as reduced MHC I tetramer binding and CD69 expression, altered distribution of lipid rafts, decreased cytolytic activity, and less susceptibility to apoptosis. Furthermore, exogenous IL-12 priming improved the intrinsic survival properties of memory CD8 T cells, leading to better protective immunity and vaccine-induced memory CD8 T cell responses. However, the experiments with IL-12p40- and IL-12Rbeta1-deficient mice showed similar levels of primary and memory CD8 T cell responses compared with wild-type mice, implying that endogenous IL-12 and/or IL-12R signaling in vivo is not critical for CD8 T cell immunity. Together, our results suggest that IL-12 can serve as an important, but dispensable regulatory factor for the development of CD8 T cells, and IL-12 priming could be useful in many medical applications.
Collapse
Affiliation(s)
- Jun Chang
- Division of Molecular and Life Science, National Laboratory of DNA Medicine, Pohang University of Science and Technology, Hyoja-Dong, Pohang, Kyungbuk, Republic of Korea
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
The alloimmune response can be divided into specific junctures where critical decisions between tolerance and immunity are made which define the outcome of the transplant. At these "decision nodes" various cytokines direct alloresponsive T cells to develop either a proinflammatory response aimed at graft destruction or an immunoregulatory response facilitating graft acceptance. This review will focus on the role of these cytokines in influencing the progression of an alloimmune response leading ultimately to either allograft survival or rejection.
Collapse
Affiliation(s)
- Patrick T. Walsh
- University of Pennsylvania, 700 Clinical Research Building, 415 Curie Boulevard, Philadelphia, Pennsylvania 19104
| | - Terry B. Strom
- Department of Medicine, Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215
| | - Laurence A. Turka
- University of Pennsylvania, 700 Clinical Research Building, 415 Curie Boulevard, Philadelphia, Pennsylvania 19104
- Correspondence:
| |
Collapse
|