1
|
Gul A, Pewe LL, Willems P, Mayer R, Thery F, Asselman C, Aernout I, Verbeke R, Eggermont D, Van Moortel L, Upton E, Zhang Y, Boucher K, Miret-Casals L, Demol H, De Smedt SC, Lentacker I, Radoshevich L, Harty JT, Impens F. Immunopeptidomics Mapping of Listeria monocytogenes T Cell Epitopes in Mice. Mol Cell Proteomics 2024; 23:100829. [PMID: 39147027 PMCID: PMC11414675 DOI: 10.1016/j.mcpro.2024.100829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 07/21/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024] Open
Abstract
Listeria monocytogenes is a foodborne intracellular bacterial model pathogen. Protective immunity against Listeria depends on an effective CD8+ T cell response, but very few T cell epitopes are known in mice as a common animal infection model for listeriosis. To identify epitopes, we screened for Listeria immunopeptides presented in the spleen of infected mice by mass spectrometry-based immunopeptidomics. We mapped more than 6000 mouse self-peptides presented on MHC class I molecules, including 12 high confident Listeria peptides from 12 different bacterial proteins. Bacterial immunopeptides with confirmed fragmentation spectra were further tested for their potential to activate CD8+ T cells, revealing VTYNYINI from the putative cell wall surface anchor family protein LMON_0576 as a novel bona fide peptide epitope. The epitope showed high biological potency in a prime boost model and can be used as a research tool to probe CD8+ T cell responses in the mouse models of Listeria infection. Together, our results demonstrate the power of immunopeptidomics for bacterial antigen identification.
Collapse
Affiliation(s)
- Adillah Gul
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Lecia L Pewe
- Department of Pathology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA
| | - Patrick Willems
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB-UGent Center for Plant Systems Biology, VIB, Ghent, Belgium; Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
| | - Rupert Mayer
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium
| | - Fabien Thery
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Caroline Asselman
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Ilke Aernout
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Rein Verbeke
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Denzel Eggermont
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Laura Van Moortel
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Ellen Upton
- Department of Microbiology and Immunology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA
| | - Yifeng Zhang
- Department of Microbiology and Immunology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA
| | - Katie Boucher
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium
| | - Laia Miret-Casals
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Hans Demol
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium
| | - Stefaan C De Smedt
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Ine Lentacker
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Lilliana Radoshevich
- Department of Microbiology and Immunology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA; Department of Immunology and Genomic Medicine, National Jewish Health, Denver, Colorado, USA.
| | - John T Harty
- Department of Pathology, University of Iowa-Carver College of Medicine, Iowa City, Iowa, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, USA.
| | - Francis Impens
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent, Belgium; VIB Proteomics Core, VIB, Ghent, Belgium.
| |
Collapse
|
2
|
Yang C, Liu Z, Yang Y, Cocka LJ, Li Y, Zeng W, Shen H. Chronic viral infection impairs immune memory to a different pathogen. PLoS Pathog 2024; 20:e1012113. [PMID: 38547316 PMCID: PMC11003680 DOI: 10.1371/journal.ppat.1012113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 04/09/2024] [Accepted: 03/11/2024] [Indexed: 04/11/2024] Open
Abstract
Chronic viral infections cause T cell dysfunction in both animal models and human clinical settings, thereby affecting the ability of the host immune system to clear viral pathogens and develop proper virus-specific immune memory. However, the impact of chronic viral infections on the host's immune memory to other pathogens has not been well described. In this study, we immunized mice with recombinant Listeria monocytogenes expressing OVA (Lm-OVA) to generate immunity to Lm and allow analysis of OVA-specific memory T (Tm) cells. We then infected these mice with lymphocytic choriomeningitis virus (LCMV) strain Cl-13 which establishes a chronic infection. We found that chronically infected mice were unable to protect against Listeria re-challenge. OVA-specific Tm cells showed a progressive loss in total numbers and in their ability to produce effector cytokines in the context of chronic LCMV infection. Unlike virus-specific T cells, OVA-specific Tm cells from chronically infected mice did not up-regulate the expression of inhibitory receptors, a hallmark feature of exhaustion in virus-specific T cells. Finally, OVA-specific Tm cells failed to mount a robust recall response after bacteria re-challenge both in the chronically infected and adoptively transferred naïve hosts. These results show that previously established bacteria-specific Tm cells become functionally impaired in the setting of an unrelated bystander chronic viral infection, which may contribute to poor immunity against other pathogens in the host with chronic viral infection.
Collapse
Affiliation(s)
- Cheng Yang
- Department of Infectious Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Pennsylvania, Philadelphia, United States of America
| | - Zhicui Liu
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Pennsylvania, Philadelphia, United States of America
- Department of Dermatology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ying Yang
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Pennsylvania, Philadelphia, United States of America
- Hainan Academy of Medical Sciences, Hainan Medical University, Hainan, China
| | - Luis J. Cocka
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Pennsylvania, Philadelphia, United States of America
| | - Yongguo Li
- Department of Infectious Diseases, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Weihong Zeng
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Pennsylvania, Philadelphia, United States of America
- Shanghai Key Laboratory of Embryo Original Diseases, the International Peace Maternity & Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Shen
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Pennsylvania, Philadelphia, United States of America
| |
Collapse
|
3
|
Wells AC, Hioki KA, Angelou CC, Lynch AC, Liang X, Ryan DJ, Thesmar I, Zhanybekova S, Zuklys S, Ullom J, Cheong A, Mager J, Hollander GA, Pobezinskaya EL, Pobezinsky LA. Let-7 enhances murine anti-tumor CD8 T cell responses by promoting memory and antagonizing terminal differentiation. Nat Commun 2023; 14:5585. [PMID: 37696797 PMCID: PMC10495470 DOI: 10.1038/s41467-023-40959-7] [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: 12/21/2022] [Accepted: 08/17/2023] [Indexed: 09/13/2023] Open
Abstract
The success of the CD8 T cell-mediated immune response against infections and tumors depends on the formation of a long-lived memory pool, and the protection of effector cells from exhaustion. The advent of checkpoint blockade therapy has significantly improved anti-tumor therapeutic outcomes by reversing CD8 T cell exhaustion, but fails to generate effector cells with memory potential. Here, using in vivo mouse models, we show that let-7 miRNAs determine CD8 T cell fate, where maintenance of let-7 expression during early cell activation results in memory CD8 T cell formation and tumor clearance. Conversely, let-7-deficiency promotes the generation of a terminal effector population that becomes vulnerable to exhaustion and cell death in immunosuppressive environments and fails to reject tumors. Mechanistically, let-7 restrains metabolic changes that occur during T cell activation through the inhibition of the PI3K/AKT/mTOR signaling pathway and production of reactive oxygen species, potent drivers of terminal differentiation and exhaustion. Thus, our results reveal a role for let-7 in the time-sensitive support of memory formation and the protection of effector cells from exhaustion. Overall, our data suggest a strategy in developing next-generation immunotherapies by preserving the multipotency of effector cells rather than enhancing the efficacy of differentiation.
Collapse
Affiliation(s)
- Alexandria C Wells
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Kaito A Hioki
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
- UMass Biotech Training Program (BTP), Amherst, MA, USA
| | - Constance C Angelou
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Adam C Lynch
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Xueting Liang
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Daniel J Ryan
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Iris Thesmar
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Saule Zhanybekova
- Pediatric Immunology, Department of Biomedicine, University of Basel and University Children's Hospital Basel, Basel, Switzerland
| | - Saulius Zuklys
- Pediatric Immunology, Department of Biomedicine, University of Basel and University Children's Hospital Basel, Basel, Switzerland
| | - Jacob Ullom
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Agnes Cheong
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Jesse Mager
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA
| | - Georg A Hollander
- Pediatric Immunology, Department of Biomedicine, University of Basel and University Children's Hospital Basel, Basel, Switzerland
| | - Elena L Pobezinskaya
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA.
| | - Leonid A Pobezinsky
- Department of Veterinary and Animal science, University of Massachusetts, Amherst, MA, USA.
| |
Collapse
|
4
|
Hassert M, Arumugam S, Harty JT. Memory CD8+ T cell-mediated protection against liver-stage malaria. Immunol Rev 2023; 316:84-103. [PMID: 37014087 PMCID: PMC10524177 DOI: 10.1111/imr.13202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/09/2023] [Accepted: 03/21/2023] [Indexed: 04/05/2023]
Abstract
Nearly half of the world's population is at risk of malaria, a disease caused by the protozoan parasite Plasmodium, which is estimated to cause more than 240,000,000 infections and kill more than 600,000 people annually. The emergence of Plasmodia resistant to chemoprophylactic treatment highlights the urgency to develop more effective vaccines. In this regard, whole sporozoite vaccination approaches in murine models and human challenge studies have provided substantial insight into the immune correlates of protection from malaria. From these studies, CD8+ T cells have come to the forefront, being identified as critical for vaccine-mediated liver-stage immunity that can prevent the establishment of the symptomatic blood stages and subsequent transmission of infection. However, the unique biological characteristics required for CD8+ T cell protection from liver-stage malaria dictate that more work must be done to design effective vaccines. In this review, we will highlight a subset of studies that reveal basic aspects of memory CD8+ T cell-mediated protection from liver-stage malaria infection.
Collapse
Affiliation(s)
- Mariah Hassert
- Department of Pathology, University of Iowa- Carver College of Medicine, Iowa City, IA, USA
| | - Sahaana Arumugam
- Department of Pathology, University of Iowa- Carver College of Medicine, Iowa City, IA, USA
- Medical Scientist Training Program, University of Iowa- Carver College of Medicine, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Immunology, University of Iowa- Carver College of Medicine, Iowa City, IA, USA
| | - John T. Harty
- Department of Pathology, University of Iowa- Carver College of Medicine, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Immunology, University of Iowa- Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
5
|
Leddy O, White FM, Bryson BD. Immunopeptidomics reveals determinants of Mycobacterium tuberculosis antigen presentation on MHC class I. eLife 2023; 12:e84070. [PMID: 37073954 PMCID: PMC10159623 DOI: 10.7554/elife.84070] [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: 10/10/2022] [Accepted: 04/17/2023] [Indexed: 04/20/2023] Open
Abstract
CD8+ T cell recognition of Mycobacterium tuberculosis (Mtb)-specific peptides presented on major histocompatibility complex class I (MHC-I) contributes to immunity to tuberculosis (TB), but the principles that govern presentation of Mtb antigens on MHC-I are incompletely understood. In this study, mass spectrometry (MS) analysis of the MHC-I repertoire of Mtb-infected primary human macrophages reveals that substrates of Mtb's type VII secretion systems (T7SS) are overrepresented among Mtb-derived peptides presented on MHC-I. Quantitative, targeted MS shows that ESX-1 activity is required for presentation of Mtb peptides derived from both ESX-1 substrates and ESX-5 substrates on MHC-I, consistent with a model in which proteins secreted by multiple T7SSs access a cytosolic antigen processing pathway via ESX-1-mediated phagosome permeabilization. Chemical inhibition of proteasome activity, lysosomal acidification, or cysteine cathepsin activity did not block presentation of Mtb antigens on MHC-I, suggesting involvement of other proteolytic pathways or redundancy among multiple pathways. Our study identifies Mtb antigens presented on MHC-I that could serve as targets for TB vaccines, and reveals how the activity of multiple T7SSs interacts to contribute to presentation of Mtb antigens on MHC-I.
Collapse
Affiliation(s)
- Owen Leddy
- Department of Biological Engineering, Massachusetts Institute of TechnologyCambridgeUnited States
- Ragon Institute of Massachusetts General Hospital, Harvard, and MITCambridgeUnited States
- Koch Institute for Integrative Cancer ResearchCambridgeUnited States
| | - Forest M White
- Department of Biological Engineering, Massachusetts Institute of TechnologyCambridgeUnited States
- Koch Institute for Integrative Cancer ResearchCambridgeUnited States
- Center for Precision Cancer MedicineCambridgeUnited States
| | - Bryan D Bryson
- Department of Biological Engineering, Massachusetts Institute of TechnologyCambridgeUnited States
- Ragon Institute of Massachusetts General Hospital, Harvard, and MITCambridgeUnited States
| |
Collapse
|
6
|
von Hoesslin M, Kuhlmann M, de Almeida GP, Kanev K, Wurmser C, Gerullis AK, Roelli P, Berner J, Zehn D. Secondary infections rejuvenate the intestinal CD103
+
tissue-resident memory T cell pool. Sci Immunol 2022; 7:eabp9553. [DOI: 10.1126/sciimmunol.abp9553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Resident T lymphocytes (T
RM
) protect tissues during pathogen reexposure. Although T
RM
phenotype and restricted migratory pattern are established, we have a limited understanding of their response kinetics, stability, and turnover during reinfections. Such characterizations have been restricted by the absence of in vivo fate-mapping systems. We generated two mouse models, one to stably mark CD103
+
T cells (a marker of T
RM
cells) and the other to specifically deplete CD103
−
T cells. Using these models, we observed that intestinal CD103
+
T cells became activated during viral or bacterial reinfection, remained organ-confined, and retained their original phenotype but failed to reexpand. Instead, the population was largely rejuvenated by CD103
+
T cells formed de novo during reinfections. This pattern remained unchanged upon deletion of antigen-specific circulating T cells, indicating that the lack of expansion was not due to competition with circulating subsets. Thus, although intestinal CD103
+
resident T cells survived long term without antigen, they lacked the ability of classical memory T cells to reexpand. This indicated that CD103
+
T cell populations could not autonomously maintain themselves. Instead, their numbers were sustained during reinfection via de novo formation from CD103
−
precursors. Moreover, in contrast to CD103
-
cells, which require antigen plus inflammation for their activation, CD103
+
T
RM
became fully activated follwing exposure to inflammation alone. Together, our data indicate that primary CD103
+
resident memory T cells lack secondary expansion potential and require CD103
−
precursors for their long-term maintenance.
Collapse
Affiliation(s)
- Madlaina von Hoesslin
- Division of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Miriam Kuhlmann
- Division of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Gustavo Pereira de Almeida
- Division of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Kristiyan Kanev
- Division of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Christine Wurmser
- Division of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Ann-Katrin Gerullis
- Division of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | | | - Jacqueline Berner
- Division of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Dietmar Zehn
- Division of Animal Physiology and Immunology, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| |
Collapse
|
7
|
Sheldon RD, Ma EH, DeCamp LM, Williams KS, Jones RG. Interrogating in vivo T-cell metabolism in mice using stable isotope labeling metabolomics and rapid cell sorting. Nat Protoc 2021; 16:4494-4521. [PMID: 34349284 DOI: 10.1038/s41596-021-00586-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 05/31/2021] [Indexed: 11/10/2022]
Abstract
T cells are integral players in the adaptive immune system that readily adapt their metabolism to meet their energetic and biosynthetic needs. A major hurdle to understand physiologic T-cell metabolism has been the differences between in vitro cell culture conditions and the complex in vivo milieu. To address this, we have developed a protocol that merges traditional immunology infection models with whole-body metabolite infusion and mass-spectrometry-based metabolomic profiling to assess T-cell metabolism in vivo. In this protocol, pathogen-infected mice are infused via the tail vein with an isotopically labeled metabolite (2-6 h), followed by rapid magnetic bead isolation to purify T-cell populations (<1 h) and then stable isotope labeling analysis conducted by mass spectrometry (~1-2 d). This procedure enables researchers to evaluate metabolic substrate utilization into central carbon metabolic pathways (i.e., glycolysis and the tricarboxylic acid cycle) by specific T-cell subpopulations in the context of physiological immune responses in vivo.
Collapse
Affiliation(s)
- Ryan D Sheldon
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA.,Metabolomics and Bioenergetics Core Facility, Van Andel Institute, Grand Rapids, MI, USA
| | - Eric H Ma
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Lisa M DeCamp
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Kelsey S Williams
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Russell G Jones
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA.
| |
Collapse
|
8
|
Chávez-Arroyo A, Portnoy DA. Why is Listeria monocytogenes such a potent inducer of CD8+ T-cells? Cell Microbiol 2021; 22:e13175. [PMID: 32185899 DOI: 10.1111/cmi.13175] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/21/2020] [Accepted: 01/22/2020] [Indexed: 12/20/2022]
Abstract
Listeria monocytogenes is a rapidly growing, Gram-positive, facultative intracellular pathogen that has been used for over 5 decades as a model to study basic aspects of infection and immunity. In a murine intravenous infection model, immunisation with a sublethal infection of L. monocytogenes initially leads to rapid intracellular multiplication followed by clearance of the bacteria and ultimately culminates in the development of long-lived cell-mediated immunity (CMI) mediated by antigen-specific CD8+ cytotoxic T-cells. Importantly, effective immunisation requires live, replicating bacteria. In this review, we summarise the cell and immunobiology of L. monocytogenes infection and discuss aspects of its pathogenesis that we suspect lead to robust CMI. We suggest five specific features of L. monocytogenes infection that positively impact the development of CMI: (a) the bacteria have a predilection for professional antigen-presenting cells; (b) the bacteria escape from phagosomes, grow, and secrete antigens into the host cell cytosol; (c) bacterial-secreted proteins enter the major histocompatibility complex (MHC) class I pathway of antigen processing and presentation; (d) the bacteria do not induce rapid host cell death; and (e) cytosolic bacteria induce a cytokine response that favours CMI. Collectively, these features make L. monocytogenes an attractive vaccine vector for both infectious disease applications and cancer immunotherapy.
Collapse
Affiliation(s)
- Alfredo Chávez-Arroyo
- Graduate Group in Microbiology, University of California, Berkeley, Berkeley, California
| | - Daniel A Portnoy
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California.,Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley, California
| |
Collapse
|
9
|
Levine LS, Hiam-Galvez KJ, Marquez DM, Tenvooren I, Madden MZ, Contreras DC, Dahunsi DO, Irish JM, Oluwole OO, Rathmell JC, Spitzer MH. Single-cell analysis by mass cytometry reveals metabolic states of early-activated CD8 + T cells during the primary immune response. Immunity 2021; 54:829-844.e5. [PMID: 33705706 PMCID: PMC8046726 DOI: 10.1016/j.immuni.2021.02.018] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 12/08/2020] [Accepted: 02/17/2021] [Indexed: 02/08/2023]
Abstract
Memory T cells are thought to rely on oxidative phosphorylation and short-lived effector T cells on glycolysis. Here, we investigated how T cells arrive at these states during an immune response. To understand the metabolic state of rare, early-activated T cells, we adapted mass cytometry to quantify metabolic regulators at single-cell resolution in parallel with cell signaling, proliferation, and effector function. We interrogated CD8+ T cell activation in vitro and in response to Listeria monocytogenes infection in vivo. This approach revealed a distinct metabolic state in early-activated T cells characterized by maximal expression of glycolytic and oxidative metabolic proteins. Cells in this transient state were most abundant 5 days post-infection before rapidly decreasing metabolic protein expression. Analogous findings were observed in chimeric antigen receptor (CAR) T cells interrogated longitudinally in advanced lymphoma patients. Our study demonstrates the utility of single-cell metabolic analysis by mass cytometry to identify metabolic adaptations of immune cell populations in vivo and provides a resource for investigations of metabolic regulation of immune responses across a variety of applications.
Collapse
Affiliation(s)
- Lauren S Levine
- Departments of Otolaryngology-Head and Neck Cancer, University of California, San Francisco, San Francisco, CA 94143, USA; G.W. Hooper Research Foundation, Department of Immunology and Microbiology, University of California, San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kamir J Hiam-Galvez
- Departments of Otolaryngology-Head and Neck Cancer, University of California, San Francisco, San Francisco, CA 94143, USA; G.W. Hooper Research Foundation, Department of Immunology and Microbiology, University of California, San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA
| | - Diana M Marquez
- Departments of Otolaryngology-Head and Neck Cancer, University of California, San Francisco, San Francisco, CA 94143, USA; G.W. Hooper Research Foundation, Department of Immunology and Microbiology, University of California, San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA
| | - Iliana Tenvooren
- Departments of Otolaryngology-Head and Neck Cancer, University of California, San Francisco, San Francisco, CA 94143, USA; G.W. Hooper Research Foundation, Department of Immunology and Microbiology, University of California, San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA
| | - Matthew Z Madden
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Diana C Contreras
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Debolanle O Dahunsi
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jonathan M Irish
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Olalekan O Oluwole
- Department of Medicine, Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jeffrey C Rathmell
- Vanderbilt Center for Immunobiology, Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Matthew H Spitzer
- Departments of Otolaryngology-Head and Neck Cancer, University of California, San Francisco, San Francisco, CA 94143, USA; G.W. Hooper Research Foundation, Department of Immunology and Microbiology, University of California, San Francisco, San Francisco, CA 94143, USA; Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA.
| |
Collapse
|
10
|
Fan Y, Bai T, Tian Y, Zhou B, Wang Y, Yang L. H 2O 2-Inactivated Salmonella typhimurium RE88 Strain as a New Cancer Vaccine Carrier: Evaluation in a Mouse Model of Cancer. Drug Des Devel Ther 2021; 15:209-222. [PMID: 33488068 PMCID: PMC7815095 DOI: 10.2147/dddt.s282660] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 12/07/2020] [Indexed: 02/05/2023] Open
Abstract
Purpose This study aimed to describe a novel cancer vaccine developed using H2O2-inactivated Salmonella typhimurium RE88 [with deletions of AroA (the first enzyme in the aromatic amino acid biosynthesis pathway) and DNA adenine methylase] as the carrier. Methods The pVLT33 plasmid was used to engineer an RE88 strain induced to express ovalbumin (OVA) by isopropylthiogalactoside (RE88-pVLT33-OVA). The immune responses and anticancer effects of H2O2-inactivated RE88-pVLT33-OVA were compared with those of non-inactivated RE88-pVLT33-OVA and OVA (positive control) in mice carrying OVA-expressing tumors (EG7-OVA) cells. Results Anti-ovalbumin IgG (immunoglobulin G) titer following vaccination with H2O2-inactivated RE88-pVLT33-OVA was higher for subcutaneous than for intragastric vaccination. When subcutaneous administration was used, H2O2-inactivated RE88-pVLT33-OVA (2 × 109 CFU (colony forming units)/mouse) achieved an anti-ovalbumin IgG titer higher than that for the same dose of RE88-pVLT33-OVA and comparable to that for 10 µg ovalbumin (positive control). The binding of mouse serum antibodies to EG7-OVA cells was stronger for H2O2-inactivated RE88-pVLT33-OVA (2 × 109 CFU/mouse) than for 10 µg ovalbumin. Furthermore, subcutaneous vaccination with H2O2-inactivated RE88-pVLT33-OVA (2 × 109 CFU/mouse) induced greater activation of splenic T cells and more extensive tumor infiltration with CD4+/CD8+ T cells compared with 10 µg ovalbumin (positive control). The mice vaccinated subcutaneously with H2O2-inactivated RE88-pVLT33-OVA at a dose of 2 × 108 or 6 × 108 CFU/mouse had smaller tumors compared with mice in the negative control groups. Tumor weight in mice vaccinated with H2O2-inactivated RE88-pVLT33-OVA at a dose of 2 × 109 CFU/mouse was significantly lower than that in both negative control groups (P < 0.05) and decreased with the increasing dose of H2O2-inactivated RE88-pVLT33-OVA. H2O2-inactivated RE88-pVLT33-OVA was potentially safer than the non-inactivated strain, could carry exogenous antigens, and had specific epitopes that could be exploited as natural adjuvants to facilitate the induction of cellular and humoral immune responses. Conclusion It was anticipated that H2O2-inactivated RE88-pVLT33-OVA could be used as a novel delivery system for new cancer vaccines.
Collapse
Affiliation(s)
- Yingzi Fan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, People's Republic of China.,Department of Laboratory Medicine, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, People's Republic of China
| | - Tingting Bai
- Department of Laboratory Medicine, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, People's Republic of China
| | - Yaomei Tian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, People's Republic of China
| | - Bailing Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, People's Republic of China
| | - Yuanda Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, People's Republic of China
| | - Li Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, People's Republic of China
| |
Collapse
|
11
|
Lee W, Kingstad-Bakke B, Paulson B, Larsen A, Overmyer K, Marinaik CB, Dulli K, Toy R, Vogel G, Mueller KP, Tweed K, Walsh AJ, Russell J, Saha K, Reyes L, Skala MC, Sauer JD, Shayakhmetov DM, Coon J, Roy K, Suresh M. Carbomer-based adjuvant elicits CD8 T-cell immunity by inducing a distinct metabolic state in cross-presenting dendritic cells. PLoS Pathog 2021; 17:e1009168. [PMID: 33444400 PMCID: PMC7840022 DOI: 10.1371/journal.ppat.1009168] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 01/27/2021] [Accepted: 11/16/2020] [Indexed: 01/25/2023] Open
Abstract
There is a critical need for adjuvants that can safely elicit potent and durable T cell-based immunity to intracellular pathogens. Here, we report that parenteral vaccination with a carbomer-based adjuvant, Adjuplex (ADJ), stimulated robust CD8 T-cell responses to subunit antigens and afforded effective immunity against respiratory challenge with a virus and a systemic intracellular bacterial infection. Studies to understand the metabolic and molecular basis for ADJ's effect on antigen cross-presentation by dendritic cells (DCs) revealed several unique and distinctive mechanisms. ADJ-stimulated DCs produced IL-1β and IL-18, suggestive of inflammasome activation, but in vivo activation of CD8 T cells was unaffected in caspase 1-deficient mice. Cross-presentation induced by TLR agonists requires a critical switch to anabolic metabolism, but ADJ enhanced cross presentation without this metabolic switch in DCs. Instead, ADJ induced in DCs, an unique metabolic state, typified by dampened oxidative phosphorylation and basal levels of glycolysis. In the absence of increased glycolytic flux, ADJ modulated multiple steps in the cytosolic pathway of cross-presentation by enabling accumulation of degraded antigen, reducing endosomal acidity and promoting antigen localization to early endosomes. Further, by increasing ROS production and lipid peroxidation, ADJ promoted antigen escape from endosomes to the cytosol for degradation by proteasomes into peptides for MHC I loading by TAP-dependent pathways. Furthermore, we found that induction of lipid bodies (LBs) and alterations in LB composition mediated by ADJ were also critical for DC cross-presentation. Collectively, our model challenges the prevailing metabolic paradigm by suggesting that DCs can perform effective DC cross-presentation, independent of glycolysis to induce robust T cell-dependent protective immunity to intracellular pathogens. These findings have strong implications in the rational development of safe and effective immune adjuvants to potentiate robust T-cell based immunity.
Collapse
Affiliation(s)
- Woojong Lee
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Brock Kingstad-Bakke
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Brett Paulson
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Autumn Larsen
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Katherine Overmyer
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Chandranaik B. Marinaik
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Kelly Dulli
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Randall Toy
- The Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University and The Parker H. Petit Institute for Bioengineering and Biosciences, Center for ImmunoEngineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Gabriela Vogel
- The Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University and The Parker H. Petit Institute for Bioengineering and Biosciences, Center for ImmunoEngineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Katherine P. Mueller
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Kelsey Tweed
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Alex J. Walsh
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Jason Russell
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Krishanu Saha
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Leticia Reyes
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Melissa C. Skala
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - John-Demian Sauer
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Dmitry M. Shayakhmetov
- Lowance Center for Human Immunology, Emory Vaccine Center, Departments of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Joshua Coon
- Morgridge Institute for Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Krishnendu Roy
- The Wallace H. Coulter Department of Biomedical Engineering at Georgia Institute of Technology and Emory University and The Parker H. Petit Institute for Bioengineering and Biosciences, Center for ImmunoEngineering, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - M. Suresh
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
12
|
Tabachnick-Cherny S, Pinto S, Berko D, Curato C, Wolf Y, Porat Z, Karmona R, Tirosh B, Jung S, Navon A. Polyglutamine-Related Aggregates Can Serve as a Potent Antigen Source for Cross-Presentation by Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2020; 205:2583-2594. [PMID: 33067378 DOI: 10.4049/jimmunol.1901535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 09/15/2020] [Indexed: 01/16/2023]
Abstract
Protective MHC class I-dependent immune responses require an overlap between repertoires of proteins directly presented on target cells and cross-presented by professional APC, specifically dendritic cells. How stable proteins that rely on defective ribosomal proteins for direct presentation are captured for cell-to-cell transfer remains enigmatic. In this study, we address this issue using a combination of in vitro (C57BL/6-derived mouse cell lines) and in vivo (C57BL/6 mouse strains) approaches involving stable and unstable versions of OVA model Ags displaying defective ribosomal protein-dependent and -independent Ag presentation, respectively. Apoptosis, but not necrosis, of donor cells was found associated with robust global protein aggregate formation and captured stable proteins permissive for cross-presentation. Potency of aggregates to serve as Ag source was directly demonstrated using polyglutamine-equipped model substrates. Collectively, our data implicate global protein aggregation in apoptotic cells as a mechanism that ensures the overlap between MHC class I epitopes presented directly or cross-presented by APC and demonstrate the unusual ability of dendritic cells to process stable protein aggregates.
Collapse
Affiliation(s)
- Shira Tabachnick-Cherny
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Sivan Pinto
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Dikla Berko
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Caterina Curato
- Department of Immunology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yochai Wolf
- Department of Immunology, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ziv Porat
- Department of Biological Services, The Weizmann Institute of Science, Rehovot 76100, Israel; and
| | - Rotem Karmona
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Boaz Tirosh
- The Institute for Drug Research, The School of Pharmacy, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Steffen Jung
- Department of Immunology, The Weizmann Institute of Science, Rehovot 76100, Israel;
| | - Ami Navon
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot 76100, Israel;
| |
Collapse
|
13
|
Pinato DJ, Guerra N, Fessas P, Murphy R, Mineo T, Mauri FA, Mukherjee SK, Thursz M, Wong CN, Sharma R, Rimassa L. Immune-based therapies for hepatocellular carcinoma. Oncogene 2020; 39:3620-3637. [PMID: 32157213 PMCID: PMC7190571 DOI: 10.1038/s41388-020-1249-9] [Citation(s) in RCA: 148] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 02/19/2020] [Accepted: 02/25/2020] [Indexed: 12/12/2022]
Abstract
Hepatocellular carcinoma (HCC) is the third most frequent cause of cancer-related death. The immune-rich contexture of the HCC microenvironment makes this tumour an appealing target for immune-based therapies. Here, we discuss how the functional characteristics of the liver microenvironment can potentially be harnessed for the treatment of HCC. We will review the evidence supporting a therapeutic role for vaccines, cell-based therapies and immune-checkpoint inhibitors and discuss the potential for patient stratification in an attempt to overcome the series of failures that has characterised drug development in this disease area.
Collapse
Affiliation(s)
- David J Pinato
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W120HS, UK.
| | - Nadia Guerra
- Department of Life Sciences, Imperial College London, South Kensington Campus, Exhibition Road, London, SW7 2AZ, UK
| | - Petros Fessas
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W120HS, UK
| | - Ravindhi Murphy
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W120HS, UK
| | | | - Francesco A Mauri
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W120HS, UK
| | - Sujit K Mukherjee
- Department of Metabolism, Digestion & Reproduction, Imperial College London, St. Mary's Hospital, Praed Street, London, UK
| | - Mark Thursz
- Department of Metabolism, Digestion & Reproduction, Imperial College London, St. Mary's Hospital, Praed Street, London, UK
| | - Ching Ngar Wong
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W120HS, UK
| | - Rohini Sharma
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W120HS, UK
| | - Lorenza Rimassa
- Medical Oncology and Haematology Unit, Humanitas Cancer Center, Humanitas Clinical and Research Center-IRCCS, Via Manzoni 56, 20089, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 20090, Pieve Emanuele, Milan, Italy
| |
Collapse
|
14
|
Morrow ZT, Powers ZM, Sauer JD. Listeria monocytogenes cancer vaccines: bridging innate and adaptive immunity. CURRENT CLINICAL MICROBIOLOGY REPORTS 2019; 6:213-224. [PMID: 33072493 DOI: 10.1007/s40588-019-00133-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Purpose of the Review Immunotherapy has emerged as a promising cancer treatment, however success in only select clinical indications underscores the need for novel approaches. Recently Listeria monocytogenes-based vaccines have been developed to drive tumor specific T-cell responses. Here, we discuss recent preclinical studies using L. monocytogenes vaccines, innate immune pathways that influence T-cell priming, and new vaccine strategies in clinical trials. Recent Findings Recent studies indicate that in addition to inducing antigen specific T-cell responses, L. monocytogenes vaccines remodel the TME. In addition, several innate immune pathways influence adaptive immune responses to L. monocytogenes and modulating these pathways holds promise to enhance anti-tumor T-cell responses. Summary The interplay between innate and adaptive immune responses to L. monocytogenes is poorly understood. Understanding these interactions will facilitate the design of better anti-cancer vaccines and improved use of combination therapies.
Collapse
Affiliation(s)
- Zachary T Morrow
- University of Wisconsin- Madison, School of Medicine and Public Health, Department of Medical Microbiology and Immunology
| | - Zachary M Powers
- University of Wisconsin- Madison, School of Medicine and Public Health, Department of Medical Microbiology and Immunology
| | - John-Demian Sauer
- University of Wisconsin-Madison, School of Medicine and Public Health, Department of Medical Microbiology and Immunology, 1550 Linden Dr. Rm 4203, Madison WI, 53706
| |
Collapse
|
15
|
D'Orazio SEF. Innate and Adaptive Immune Responses during Listeria monocytogenes Infection. Microbiol Spectr 2019; 7:10.1128/microbiolspec.gpp3-0065-2019. [PMID: 31124430 PMCID: PMC11086964 DOI: 10.1128/microbiolspec.gpp3-0065-2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Indexed: 12/15/2022] Open
Abstract
It could be argued that we understand the immune response to infection with Listeria monocytogenes better than the immunity elicited by any other bacteria. L. monocytogenes are Gram-positive bacteria that are genetically tractable and easy to cultivate in vitro, and the mouse model of intravenous (i.v.) inoculation is highly reproducible. For these reasons, immunologists frequently use the mouse model of systemic listeriosis to dissect the mechanisms used by mammalian hosts to recognize and respond to infection. This article provides an overview of what we have learned over the past few decades and is divided into three sections: "Innate Immunity" describes how the host initially detects the presence of L. monocytogenes and characterizes the soluble and cellular responses that occur during the first few days postinfection; "Adaptive Immunity" discusses the exquisitely specific T cell response that mediates complete clearance of infection and immunological memory; "Use of Attenuated Listeria as a Vaccine Vector" highlights the ways that investigators have exploited our extensive knowledge of anti-Listeria immunity to develop cancer therapeutics.
Collapse
Affiliation(s)
- Sarah E F D'Orazio
- University of Kentucky, Microbiology, Immunology & Molecular Genetics, Lexington, KY 40536-0298
| |
Collapse
|
16
|
Curato C, Bernshtein B, Zupancič E, Dufner A, Jaitin D, Giladi A, David E, Chappell-Maor L, Leshkowitz D, Knobeloch KP, Amit I, Florindo HF, Jung S. DC Respond to Cognate T Cell Interaction in the Antigen-Challenged Lymph Node. Front Immunol 2019; 10:863. [PMID: 31073301 PMCID: PMC6496461 DOI: 10.3389/fimmu.2019.00863] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/04/2019] [Indexed: 01/11/2023] Open
Abstract
Dendritic cells (DC) are unrivaled in their potential to prime naive T cells by presenting antigen and providing costimulation. DC are furthermore believed to decode antigen context by virtue of pattern recognition receptors and to polarize T cells through cytokine secretion toward distinct effector functions. Diverse polarized T helper (TH) cells have been explored in great detail. In contrast, studies of instructing DC have to date largely been restricted to in vitro settings or adoptively transferred DC. Here we report efforts to unravel the DC response to cognate T cell encounter in antigen-challenged lymph nodes (LN). Mice engrafted with antigen-specific T cells were immunized with nanoparticles (NP) entrapping adjuvants and absorbed with antigen to study the immediate DC response to T cell encounter using bulk and single cell RNA-seq profiling. NP induced robust antigen-specific TH1 cell responses with minimal bystander activation. Fluorescent-labeled NP allowed identification of antigen-carrying DC and focus on transcriptional changes in DC that encounter T cells. Our results support the existence of a bi-directional crosstalk between DC and T cells that promotes TH1 responses, including involvement of the ubiquitin-like molecule Isg15 that merits further study.
Collapse
Affiliation(s)
- Caterina Curato
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Biana Bernshtein
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eva Zupancič
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - Almut Dufner
- Medical Faculty, Institute for Neuropathology, University Freiburg, Freiburg, Germany
| | - Diego Jaitin
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Amir Giladi
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal David
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Dena Leshkowitz
- Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Klaus-Peter Knobeloch
- Medical Faculty, Institute for Neuropathology, University Freiburg, Freiburg, Germany
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Helena F Florindo
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
17
|
Mathews DV, Dong Y, Higginbotham LB, Kim SC, Breeden CP, Stobert EA, Jenkins J, Tso JY, Larsen CP, Adams AB. CD122 signaling in CD8+ memory T cells drives costimulation-independent rejection. J Clin Invest 2018; 128:4557-4572. [PMID: 30222140 PMCID: PMC6159972 DOI: 10.1172/jci95914] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 07/31/2018] [Indexed: 12/30/2022] Open
Abstract
Interrupting T cell costimulatory signals as a strategy to control undesired immune responses, such as occur in autoimmunity or transplantation, has the potential to alleviate many of the unwanted side effects associated with current immunosuppressive therapies. Belatacept, a high-affinity version of CTLA4-Ig that blocks ligand ligation to CD28, has been approved for use in kidney transplant recipients. Despite the long-term benefits associated with its use, such as improved renal function and lower cardiovascular risk, a subset of patients treated with belatacept experience elevated rates of acute T cell-mediated rejection, tempering enthusiasm for its use. Here we demonstrate that costimulation-independent T cell alloreactivity relies on signaling through CD122, the shared IL-2 and IL-15 receptor β-chain. Combined costimulatory and CD122 blockade improved survival of transplanted tissue in mice and nonhuman primates by controlling proliferation and effector function of CD8+ T cells. The high-affinity IL-2 receptor was dispensable for memory CD8+ T cell responses, whereas signaling through CD122 as a component of the high-affinity IL-15 receptor was critical for costimulation-independent memory CD8+ T cell recall, distinguishing specific roles for IL-2 and IL-15 in T cell activation. These studies outline a novel approach for clinical optimization of costimulatory blockade strategies in transplantation by targeting CD122.
Collapse
Affiliation(s)
- David V. Mathews
- Emory Transplant Center, Emory University, Atlanta, Georgia, USA
| | - Ying Dong
- Emory Transplant Center, Emory University, Atlanta, Georgia, USA
| | | | - Steven C. Kim
- Emory Transplant Center, Emory University, Atlanta, Georgia, USA
| | | | | | | | - J. Yun Tso
- JN Biosciences, Mountain View, California, USA
| | - Christian P. Larsen
- Emory Transplant Center, Emory University, Atlanta, Georgia, USA
- Yerkes National Primate Center, Atlanta, Georgia, USA
| | - Andrew B. Adams
- Emory Transplant Center, Emory University, Atlanta, Georgia, USA
- Yerkes National Primate Center, Atlanta, Georgia, USA
| |
Collapse
|
18
|
Listeria Monocytogenes: A Model Pathogen Continues to Refine Our Knowledge of the CD8 T Cell Response. Pathogens 2018; 7:pathogens7020055. [PMID: 29914156 PMCID: PMC6027175 DOI: 10.3390/pathogens7020055] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Revised: 06/13/2018] [Accepted: 06/14/2018] [Indexed: 12/12/2022] Open
Abstract
Listeria monocytogenes (Lm) infection induces robust CD8 T cell responses, which play a critical role in resolving Lm during primary infection and provide protective immunity to re-infections. Comprehensive studies have been conducted to delineate the CD8 T cell response after Lm infection. In this review, the generation of the CD8 T cell response to Lm infection will be discussed. The role of dendritic cell subsets in acquiring and presenting Lm antigens to CD8 T cells and the events that occur during T cell priming and activation will be addressed. CD8 T cell expansion, differentiation and contraction as well as the signals that regulate these processes during Lm infection will be explored. Finally, the formation of memory CD8 T cell subsets in the circulation and in the intestine will be analyzed. Recently, the study of CD8 T cell responses to Lm infection has begun to shift focus from the intravenous infection model to a natural oral infection model as the humanized mouse and murinized Lm have become readily available. Recent findings in the generation of CD8 T cell responses to oral infection using murinized Lm will be explored throughout the review. Finally, CD8 T cell-mediated protective immunity against Lm infection and the use of Lm as a vaccine vector for cancer immunotherapy will be highlighted. Overall, this review will provide detailed knowledge on the biology of CD8 T cell responses after Lm infection that may shed light on improving rational vaccine design.
Collapse
|
19
|
Jahn ML, Steffensen MA, Christensen JP, Thomsen AR. Analysis of adenovirus-induced immunity to infection with Listeria monocytogenes: Fading protection coincides with declining CD8 T cell numbers and phenotypic changes. Vaccine 2018; 36:2825-2832. [PMID: 29627230 DOI: 10.1016/j.vaccine.2018.03.080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 03/23/2018] [Accepted: 03/28/2018] [Indexed: 01/09/2023]
Abstract
Defining correlates of T cell mediated protection is important in order to accelerate the development of efficient T cell based vaccines conferring long-term immunity. Extensive studies have provided important insight regarding the characteristics and functional properties of the effector and memory CD8 T cells induced by viral vector based vaccines. However, long-term protection has been difficult to achieve with T cell inducing vaccines, and the determinants underlying this loss in protection over time are still not fully defined. In this study we analyzed different parameters of the CD8 T cell response as a function of time after vaccination with a human serotype 5 adenovector expressing the glycoprotein (GP) of LCMV tethered to the MHC class II-associated invariant chain. Using this vector we have previously found that CD8 T cells mediate protection from challenge with GP-expressing Listeria monocytogenes at 60 days post vaccination, but only little protection after further 60 days, and we now confirm this observation. A comparison of vaccine-primed CD8 T cells early and late after vaccination revealed a minor decline in the overall numbers of antigen specific memory CD8 T cells during this interval. More importantly, we also observed phenotypic changes over time with a distinct decline in the frequency and number of KLRG1+ CD8 T cells, and, notably, adoptive transfer studies confirmed that memory CD8 T cells expressing KLRG1 are central to protection from systemic L. monocytogenes infection. Together these findings imply that multiple factors including changes in memory T cell numbers and phenotypic composition over time influence the longevity of CD8 T-cell mediated protection.
Collapse
Affiliation(s)
- Marie Louise Jahn
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Allan Randrup Thomsen
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
20
|
Liu D, Badell IR, Ford ML. Selective CD28 blockade attenuates CTLA-4-dependent CD8+ memory T cell effector function and prolongs graft survival. JCI Insight 2018; 3:96378. [PMID: 29321374 DOI: 10.1172/jci.insight.96378] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 11/28/2017] [Indexed: 12/21/2022] Open
Abstract
Memory T cells pose a significant problem to successful therapeutic control of unwanted immune responses during autoimmunity and transplantation, as they are differentially controlled by cosignaling receptors such as CD28 and CTLA-4. Treatment with abatacept and belatacept impede CD28 signaling by binding to CD80 and CD86, but they also have the unintended consequence of blocking the ligands for CTLA-4, a process that may inadvertently boost effector responses. Here, we show that a potentially novel anti-CD28 domain antibody (dAb) that selectively blocks CD28 but preserves CTLA-4 coinhibition confers improved allograft survival in sensitized recipients as compared with CTLA-4 Ig. However, both CTLA-4 Ig and anti-CD28 dAb similarly and significantly reduced the accumulation of donor-reactive CD8+ memory T cells, demonstrating that regulation of the expansion of CD8+ memory T cell populations is controlled in part by CD28 signals and is not significantly impacted by CTLA-4. In contrast, selective CD28 blockade was superior to CTLA-4 Ig in inhibiting IFN-γ, TNF, and IL-2 production by CD8+ memory T cells, which in turn resulted in reduced recruitment of innate CD11b+ monocytes into allografts. Importantly, this superiority was CTLA-4 dependent, demonstrating that effector function of CD8+ memory T cells is regulated by the balance of CD28 and CTLA-4 signaling.
Collapse
|
21
|
The transcription factor Runx3 guards cytotoxic CD8 + effector T cells against deviation towards follicular helper T cell lineage. Nat Immunol 2017; 18:931-939. [PMID: 28604718 PMCID: PMC5564218 DOI: 10.1038/ni.3773] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 05/18/2017] [Indexed: 12/24/2022]
Abstract
Activated CD8+ T cells differentiate into cytotoxic effector (TEFF) cells that eliminate target cells. How TEFF cell identity is established and maintained is not fully understood. We found that Runx3 deficiency limited clonal expansion and impaired upregulation of cytotoxic molecules in TEFF cells. Runx3-deficient CD8+ TEFF cells aberrantly upregulated genes characteristic of follicular helper T (TFH) cell lineage, including Bcl6, Tcf7 and Cxcr5. Mechanistically, the Runx3-CBFβ transcription factor complex deployed H3K27me3 to Bcl6 and Tcf7 genes to suppress the TFH program. Ablating Tcf7 in Runx3-deficient CD8+ TEFF cells prevented the upregulation of TFH genes and ameliorated their defective induction of cytotoxic genes. As such, Runx3-mediated Tcf7 repression coordinately enforced acquisition of cytotoxic functions and protected the cytotoxic lineage integrity by preventing TFH-lineage deviation.
Collapse
|
22
|
Tang X, Yin G, Qin M, Tao G, Suo J, Liu X, Suo X. Transgenic Eimeria tenella as a vaccine vehicle: expressing TgSAG1 elicits protective immunity against Toxoplasma gondii infections in chickens and mice. Sci Rep 2016; 6:29379. [PMID: 27387302 PMCID: PMC4937369 DOI: 10.1038/srep29379] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 06/17/2016] [Indexed: 02/02/2023] Open
Abstract
The surface antigen 1 of Toxoplasma gondii (TgSAG1) is a major immunodominant antigen and is widely considered an ideal candidate for the development of an effective recombinant vaccine against toxoplasmosis. Eimeria tenella, an affinis apicomplexan parasite with T. gondii, is a potential vaccine vector carrying exogenous antigens that stimulates specific immune responses. Here, we engineered TgSAG1 into E. tenella and obtained a stably transfected E. tenella line (Et-TgSAG1). We found TgSAG1 localized on the cell surface of Et-TgSAG1, which is similar to its native distribution in T. gondii tachyzoites. We immunized the chickens with Et-TgSAG1 orally and detected TgSAG1-specific immune responses, which partly reduced T. gondii infection. In the mouse model, we immunized the mice with Et-TgSAG1 sporozoites intraperitoneally and challenged them with T. gondii tachyzoites RH strain. We found that the mice immunized with Et-TgSAG1 showed a TgSAG1 specific Th 1-dominant immune response and a prolonged survival time compared with wild-type E. tenella and non-immunized mice. Collectively, our results demonstrated that Et-TgSAG1, utilized as a recombinant vaccine against toxoplasmosis, could be applied in both chickens and mice. Our findings also provide a promising persuasion for the development of transgenic Eimeria as vaccine vectors for use in birds and mammals.
Collapse
Affiliation(s)
- Xinming Tang
- State Key Laboratory of Agrobiotechnology &Key Laboratory of Zoonosis of Ministry of Agriculture &National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Guangwen Yin
- Engineering Laboratory of Animal Pharmaceuticals, College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou, 350002, Fujian Province, China
| | - Mei Qin
- State Key Laboratory of Agrobiotechnology &Key Laboratory of Zoonosis of Ministry of Agriculture &National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Geru Tao
- State Key Laboratory of Agrobiotechnology &Key Laboratory of Zoonosis of Ministry of Agriculture &National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Jingxia Suo
- State Key Laboratory of Agrobiotechnology &Key Laboratory of Zoonosis of Ministry of Agriculture &National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Xianyong Liu
- State Key Laboratory of Agrobiotechnology &Key Laboratory of Zoonosis of Ministry of Agriculture &National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Xun Suo
- State Key Laboratory of Agrobiotechnology &Key Laboratory of Zoonosis of Ministry of Agriculture &National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| |
Collapse
|
23
|
Doll KL, Pewe LL, Kurup SP, Harty JT. Discriminating Protective from Nonprotective Plasmodium-Specific CD8+ T Cell Responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 196:4253-62. [PMID: 27084099 PMCID: PMC4868661 DOI: 10.4049/jimmunol.1600155] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 03/19/2016] [Indexed: 11/19/2022]
Abstract
Despite decades of research, malaria remains a global health crisis. Current subunit vaccine approaches do not provide efficient long-term, sterilizing immunity against Plasmodium infections in humans. Conversely, whole parasite vaccinations with their larger array of target Ags have conferred long-lasting sterilizing protection to humans. Similar studies in rodent models of malaria reveal that CD8(+) T cells play a critical role in liver-stage immunity after whole parasite vaccination. However, it is unknown whether all CD8(+) T cell specificities elicited by whole parasite vaccination contribute to protection, an issue of great relevance for enhanced subunit vaccination. In this article, we show that robust CD8(+) T cell responses of similar phenotype are mounted after prime-boost immunization against Plasmodium berghei glideosome-associated protein 5041-48-, sporozoite-specific protein 20318-325-, thrombospondin-related adhesion protein (TRAP) 130-138-, or circumsporozoite protein (CSP) 252-260-derived epitopes in mice, but only CSP252-260- and TRAP130-138-specific CD8(+) T cells provide sterilizing immunity and reduce liver parasite burden after sporozoite challenge. Further, CD8(+) T cells specific to sporozoite surface-expressed CSP and TRAP proteins, but not intracellular glideosome-associated protein 50 and sporozoite-specific protein 20, efficiently recognize sporozoite-infected hepatocytes in vitro. These results suggest that: 1) protection-relevant antigenic targets, regardless of their immunogenic potential, must be efficiently presented by infected hepatocytes for CD8(+) T cells to eliminate liver-stage Plasmodium infection; and 2) proteins expressed on the surface of sporozoites may be good target Ags for protective CD8(+) T cells.
Collapse
Affiliation(s)
- Katherine L Doll
- Department of Microbiology, University of Iowa, Iowa City, IA 52242
| | - Lecia L Pewe
- Department of Microbiology, University of Iowa, Iowa City, IA 52242
| | | | - John T Harty
- Department of Microbiology, University of Iowa, Iowa City, IA 52242; Department of Pathology, University of Iowa, Iowa City, IA 52242; and Interdisciplinary Program in Immunology, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
24
|
Liu D, Burd EM, Coopersmith CM, Ford ML. Retrogenic ICOS Expression Increases Differentiation of KLRG-1hiCD127loCD8+ T Cells during Listeria Infection and Diminishes Recall Responses. THE JOURNAL OF IMMUNOLOGY 2016; 196:1000-12. [PMID: 26729800 DOI: 10.4049/jimmunol.1500218] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 12/02/2015] [Indexed: 12/26/2022]
Abstract
Following T cell encounter with Ag, multiple signals are integrated to collectively induce distinct differentiation programs within Ag-specific CD8(+) T cell populations. Several factors contribute to these cell fate decisions, including the amount and duration of Ag, exposure to inflammatory cytokines, and degree of ligation of cosignaling molecules. The ICOS is not expressed on resting T cells but is rapidly upregulated upon encounter with Ag. However, the impact of ICOS signaling on programmed differentiation is not well understood. In this study, we therefore sought to determine the role of ICOS signaling on CD8(+) T cell programmed differentiation. Through the creation of novel ICOS retrogenic Ag-specific TCR-transgenic CD8(+) T cells, we interrogated the phenotype, functionality, and recall potential of CD8(+) T cells that receive early and sustained ICOS signaling during Ag exposure. Our results reveal that these ICOS signals critically impacted cell fate decisions of Ag-specific CD8(+) T cells, resulting in increased frequencies of KLRG-1(hi)CD127(lo) cells, altered BLIMP-1, T-bet, and eomesodermin expression, and increased cytolytic capacity as compared with empty vector controls. Interestingly, however, ICOS retrogenic CD8(+) T cells also preferentially homed to nonlymphoid organs and exhibited reduced multicytokine functionality and reduced ability to mount secondary recall responses upon challenge in vivo. In sum, our results suggest that an altered differentiation program is induced following early and sustained ICOS expression, resulting in the generation of more cytolyticly potent, terminally differentiated effectors that possess limited capacity for recall response.
Collapse
Affiliation(s)
- Danya Liu
- Emory Transplant Center, Emory University, Atlanta, GA 30322; Department of Surgery, Emory University, Atlanta, GA 30322
| | - Eileen M Burd
- Department of Pathology, Emory University, Atlanta, GA 30322; and
| | - Craig M Coopersmith
- Department of Surgery, Emory University, Atlanta, GA 30322; Emory Critical Care Center, Emory University, Atlanta, GA 30322
| | - Mandy L Ford
- Emory Transplant Center, Emory University, Atlanta, GA 30322;
| |
Collapse
|
25
|
Condotta SA, Khan SH, Rai D, Griffith TS, Badovinac VP. Polymicrobial Sepsis Increases Susceptibility to Chronic Viral Infection and Exacerbates CD8+ T Cell Exhaustion. THE JOURNAL OF IMMUNOLOGY 2015; 195:116-25. [PMID: 25980007 DOI: 10.4049/jimmunol.1402473] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 04/21/2015] [Indexed: 02/06/2023]
Abstract
Patients who survive sepsis display suppressed immune functions, often manifested as an increased susceptibility to secondary infections. Recently, using a cecal-ligation and puncture (CLP) model of sepsis, we showed that sepsis induces substantial and long-lasting changes in the available naive CD8(+) T cell repertoire affecting the capacity of the host to respond to newly encountered acute infections. However, the extent to which sepsis changes the host susceptibility to chronic infection and affects CD8(+) T cell responses is currently unknown. In this study, we demonstrate that inbred and outbred mice recovering from a septic event are more susceptible to lymphocytic choriomeningitis virus (LCMV) clone-13 infection exhibited by mortality and viral burden. Primary virus-specific CD8(+) T cells in LCMV clone-13-infected septic mice displayed exacerbated CD8(+) T cell exhaustion illustrated by increased inhibitory molecule expression (e.g., programmed cell death 1, lymphocyte-activation gene 3, and 2B4) and diminished Ag-driven cytokine production (e.g., IFN-γ, TNF-α) compared with similarly infected sham-treated mice. Importantly, therapeutic inhibitory molecule dual blockade (anti-PD-L1 and anti-lymphocyte-activation gene 3) increased the number of circulating LCMV-specific CD8(+) T cells, and improved CD8(+) T cell function and pathogen control in chronically infected septic mice. Together, these results illustrate that polymicrobial sepsis compromises the overall health of the host leading to increased vulnerability to chronic infection and exacerbated CD8(+) T cell exhaustion. Collectively, our findings suggest that septic survivors may be more susceptible and at greater risk for developing exhaustible CD8(+) T cells upon encountering a subsequent chronic infection.
Collapse
Affiliation(s)
- Stephanie A Condotta
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242
| | - Shaniya H Khan
- Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242
| | - Deepa Rai
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242
| | - Thomas S Griffith
- Microbiology, Immunology and Cancer Biology Graduate Program, University of Minnesota Medical School, Minneapolis, MN 55455; Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; Masonic Cancer Center, University of Minnesota Medical School, Minneapolis, MN 55455; Department of Urology, University of Minnesota Medical School, Minneapolis, MN 55455; and Minneapolis VA Health Care System, Minneapolis, MN 55417
| | - Vladimir P Badovinac
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, IA 52242; Interdisciplinary Program in Immunology, University of Iowa Carver College of Medicine, Iowa City, IA 52242;
| |
Collapse
|
26
|
Listeria monocytogenes: a model pathogen to study antigen-specific memory CD8 T cell responses. Semin Immunopathol 2015; 37:301-10. [PMID: 25860798 DOI: 10.1007/s00281-015-0477-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 03/02/2015] [Indexed: 10/23/2022]
Abstract
Memory CD8 T cells play a critical role in providing protection to immune hosts by orchestrating rapid elimination of pathogen-infected cells after re-infection. Systemic bacterial infection with Listeria monocytogenes has been a favored approach for researchers to characterize pathogen-specific CD8 T cell responses, and in-depth understanding of L. monocytogenes biology has provided invaluable experimental tools that have been used to increase our understanding of memory CD8 T cell differentiation. Here, we describe how the tools from this murine model system of infection have been utilized to characterize pathogen-specific CD8 T cells in inbred and genetically diverse outbred hosts as they undergo naïve-to-memory CD8 T cell differentiation in vivo. We also discuss how studying L. monocytogenes-evoked CD8 T cell responses have provided insight on the degree of diminished T cell immunity in clinically relevant conditions such as sepsis and obesity. Overall, this review will highlight how infection with the intracellular pathogen L. monocytogenes has enabled analysis of systemic CD8 T cell responses and greatly contributed to what is known about memory CD8 T cell generation and differentiation.
Collapse
|
27
|
Lee H, Kim BJ, Kim BR, Kook YH, Kim BJ. The development of a novel Mycobacterium-Escherichia coli shuttle vector system using pMyong2, a linear plasmid from Mycobacterium yongonense DSM 45126T. PLoS One 2015; 10:e0122897. [PMID: 25822634 PMCID: PMC4378964 DOI: 10.1371/journal.pone.0122897] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 02/24/2015] [Indexed: 11/18/2022] Open
Abstract
The Mycobacterium-Escherichia coli shuttle vector system, equipped with the pAL5000 replicon, is widely used for heterologous gene expression and gene delivery in mycobacteria. Despite its extensive use, this system has certain limitations, which has led to the development of alternative mycobacterial vector systems. The present study describes the molecular structure and expression profiles of a novel 18-kb linear plasmid, pMyong2, from Mycobacterium yongonense. Sixteen open reading frames and a putative origin of replication were identified, and the compatibility of the pMyong2 and pAL5000 vector systems was demonstrated. In recombinant Mycobacterium smegmatis (rSmeg), the pMyong2 vector system showed a copy number that was approximately 37 times greater than that of pAL5000. Furthermore, pMyong2 increased the mRNA and protein expression of the human macrophage migration inhibitory factor (hMIF) over pAL5000 levels by approximately 10-fold and 50-fold, respectively, demonstrating the potential utility of the pMyong2 vector system in heterologous gene expression in mycobacteria. Successful delivery of the EGFP gene into mammalian cells via rSmeg carrying the pMyong2 vector system was also observed, demonstrating the feasibility of this system for DNA delivery. In conclusion, the pMyong2 vector system could be effectively used not only for the in vivo delivery of recombinant protein and DNA but also for mycobacterial genetic studies as an alternative or a complement to the pAL5000 vector system.
Collapse
Affiliation(s)
- Hyungki Lee
- Department of Microbiology and Immunology, Biomedical Sciences, Liver Research Institute and Cancer Research Institute, College of Medicine, Seoul National University, Seoul, Korea
| | - Byoung-Jun Kim
- Department of Microbiology and Immunology, Biomedical Sciences, Liver Research Institute and Cancer Research Institute, College of Medicine, Seoul National University, Seoul, Korea
| | - Bo-Ram Kim
- Department of Microbiology and Immunology, Biomedical Sciences, Liver Research Institute and Cancer Research Institute, College of Medicine, Seoul National University, Seoul, Korea
| | - Yoon-Hoh Kook
- Department of Microbiology and Immunology, Biomedical Sciences, Liver Research Institute and Cancer Research Institute, College of Medicine, Seoul National University, Seoul, Korea
| | - Bum-Joon Kim
- Department of Microbiology and Immunology, Biomedical Sciences, Liver Research Institute and Cancer Research Institute, College of Medicine, Seoul National University, Seoul, Korea
- * E-mail:
| |
Collapse
|
28
|
Chen Z, Ozbun L, Chong N, Wallecha A, Berzofsky JA, Khleif SN. Episomal expression of truncated listeriolysin O in LmddA-LLO-E7 vaccine enhances antitumor efficacy by preferentially inducing expansions of CD4+FoxP3- and CD8+ T cells. Cancer Immunol Res 2014; 2:911-22. [PMID: 24872025 DOI: 10.1158/2326-6066.cir-13-0197] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Studies have shown that Listeria monocytogenes (Lm)-based vaccine expressing a fusion protein comprising truncated listeriolysin O (LLO) and human papilloma virus (HPV) E7 protein (Lm-LLO-E7) induces a decrease in regulatory T cells (Treg) and complete regression of established, transplanted HPV-TC-1 tumors in mice. However, how the Lm-based vaccine causes a decrease in Tregs remains unclear. Using a highly attenuated Lm dal dat ΔactA strain (LmddA)-based vaccine, we report here that the vector LmddA was sufficient to induce a decrease in the proportion of Tregs by preferentially expanding CD4(+)FoxP3(-) T cells and CD8(+) T cells by a mechanism dependent on and directly mediated by LLO. Episomal expression of a nonhemolytic truncated LLO in Lm (LmddA-LLO) significantly augmented the expansion, thus further decreasing Treg frequency. Although adoptive transfer of Tregs compromised the antitumor efficacy of the LmddA-LLO-E7 vaccine, a combination of LmddA-LLO and an Lm-based vaccine expressing E7 protein (Lm-E7) induced complete regression against established TC-1 tumors. An engineered LLO-minus Lm expressing perfringolysin O (PFO) that enables the recombinant bacteria to exit from the phagolysosome without LLO confirmed that the adjuvant effect was dependent on LLO. These results suggest that LLO may serve as a promising adjuvant by preferentially inducing the expansions of CD4(+)FoxP3(-) T cells and CD8(+) T cells, thus reducing the ratio of Tregs to CD4(+)FoxP3(-) T cells and to CD8(+) T cells favoring immune responses to eradicate tumor.
Collapse
Affiliation(s)
- Zhisong Chen
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Laurent Ozbun
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Namju Chong
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | | | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland;
| | - Samir N Khleif
- GRU Cancer Center, Georgia Regents University, Augusta, Georgia
| |
Collapse
|
29
|
Grover HS, Chu HH, Kelly FD, Yang SJ, Reese ML, Blanchard N, Gonzalez F, Chan SW, Boothroyd JC, Shastri N, Robey EA. Impact of regulated secretion on antiparasitic CD8 T cell responses. Cell Rep 2014; 7:1716-1728. [PMID: 24857659 DOI: 10.1016/j.celrep.2014.04.031] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 02/21/2014] [Accepted: 04/16/2014] [Indexed: 10/25/2022] Open
Abstract
CD8 T cells play a key role in defense against the intracellular parasite Toxoplasma, but why certain CD8 responses are more potent than others is not well understood. Here, we describe a parasite antigen, ROP5, that elicits a CD8 T cell response in genetically susceptible mice. ROP5 is secreted via parasite organelles termed rhoptries that are injected directly into host cells during invasion, whereas the protective, dense-granule antigen GRA6 is constitutively secreted into the parasitophorous vacuole. Transgenic parasites in which the ROP5 antigenic epitope was targeted for secretion through dense granules led to enhanced CD8 T cell responses, whereas targeting the GRA6 epitope to rhoptries led to reduced CD8 responses. CD8 T cell responses to the dense-granule-targeted ROP5 epitope resulted in reduced parasite load in the brain. These data suggest that the mode of secretion affects the efficacy of parasite-specific CD8 T cell responses.
Collapse
Affiliation(s)
- Harshita Satija Grover
- Division of Immunology and Pathogenesis, Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley, CA 94720-3200, USA
| | - H Hamlet Chu
- Division of Immunology and Pathogenesis, Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley, CA 94720-3200, USA
| | - Felice D Kelly
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305-5124, USA
| | - Soo Jung Yang
- Division of Immunology and Pathogenesis, Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley, CA 94720-3200, USA
| | - Michael L Reese
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305-5124, USA
| | - Nicolas Blanchard
- Center of Pathophysiology of Toulouse-Purpan, INSERM UMR1043-CNRS UMR5282, University of Toulouse, 31024 Toulouse Cedex 3, France
| | - Federico Gonzalez
- Division of Immunology and Pathogenesis, Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley, CA 94720-3200, USA
| | - Shiao Wei Chan
- Division of Immunology and Pathogenesis, Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley, CA 94720-3200, USA
| | - John C Boothroyd
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305-5124, USA
| | - Nilabh Shastri
- Division of Immunology and Pathogenesis, Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley, CA 94720-3200, USA.
| | - Ellen A Robey
- Division of Immunology and Pathogenesis, Department of Molecular and Cellular Biology, University of California, Berkeley, Berkeley, CA 94720-3200, USA.
| |
Collapse
|
30
|
Mittal R, Wagener M, Breed ER, Liang Z, Yoseph BP, Burd EM, Farris AB, Coopersmith CM, Ford ML. Phenotypic T cell exhaustion in a murine model of bacterial infection in the setting of pre-existing malignancy. PLoS One 2014; 9:e93523. [PMID: 24796533 PMCID: PMC4010417 DOI: 10.1371/journal.pone.0093523] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 03/06/2014] [Indexed: 12/22/2022] Open
Abstract
While much of cancer immunology research has focused on anti-tumor immunity both systemically and within the tumor microenvironment, little is known about the impact of pre-existing malignancy on pathogen-specific immune responses. Here, we sought to characterize the antigen-specific CD8+ T cell response following a bacterial infection in the setting of pre-existing pancreatic adenocarcinoma. Mice with established subcutaneous pancreatic adenocarcinomas were infected with Listeria monocytogenes, and antigen-specific CD8+ T cell responses were compared to those in control mice without cancer. While the kinetics and magnitude of antigen-specific CD8+ T cell expansion and accumulation was comparable between the cancer and non-cancer groups, bacterial antigen-specific CD8+ T cells and total CD4+ and CD8+ T cells in cancer mice exhibited increased expression of the coinhibitory receptors BTLA, PD-1, and 2B4. Furthermore, increased inhibitory receptor expression was associated with reduced IFN-γ and increased IL-2 production by bacterial antigen-specific CD8+ T cells in the cancer group. Taken together, these data suggest that cancer's immune suppressive effects are not limited to the tumor microenvironment, but that pre-existing malignancy induces phenotypic exhaustion in T cells by increasing expression of coinhibitory receptors and may impair pathogen-specific CD8+ T cell functionality and differentiation.
Collapse
Affiliation(s)
- Rohit Mittal
- Department of Surgery and Emory Center for Critical Care, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Maylene Wagener
- Department of Surgery and Emory Transplant Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Elise R. Breed
- Department of Surgery and Emory Center for Critical Care, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Zhe Liang
- Department of Surgery and Emory Center for Critical Care, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Benyam P. Yoseph
- Department of Surgery and Emory Center for Critical Care, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Eileen M. Burd
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Alton B. Farris
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Craig M. Coopersmith
- Department of Surgery and Emory Center for Critical Care, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Mandy L. Ford
- Department of Surgery and Emory Transplant Center, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
31
|
O'Brien S, Thomas RM, Wertheim GB, Zhang F, Shen H, Wells AD. Ikaros imposes a barrier to CD8+ T cell differentiation by restricting autocrine IL-2 production. THE JOURNAL OF IMMUNOLOGY 2014; 192:5118-29. [PMID: 24778448 DOI: 10.4049/jimmunol.1301992] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Naive CD4(+) T cells require signals from the TCR and CD28 to produce IL-2, expand, and differentiate. However, these same signals are not sufficient to induce autocrine IL-2 production by naive CD8(+) T cells, which require cytokines provided by other cell types to drive their differentiation. The basis for failed autocrine IL-2 production by activated CD8(+) cells is unclear. We find that Ikaros, a transcriptional repressor that silences IL-2 in anergic CD4(+) T cells, also restricts autocrine IL-2 production by CD8(+) T cells. We find that CD8(+) T cell activation in vitro in the absence of exogenous cytokines and CD4 help leads to marked induction of Ikaros, a known repressor of the Il2 gene. Naive murine CD8 T cells haplo-insufficient for Ikzf1 failed to upregulate Ikaros, produced autocrine IL-2, and differentiated in an IL-2-dependent manner into IFN-γ-producing CTLs in response to TCR/CD28 stimulation alone. Furthermore, Ikzf1 haplo-insufficient CD8(+) T cells were more effective at controlling Listeria infection and B16 melanoma growth in vivo, and they could provide help to neighboring, non-IL-2-producing cells to differentiate into IFN-γ-producing effectors. Therefore, by repressing autocrine IL-2 production, Ikaros ensures that naive CD8(+) T cells remain dependent on licensing by APCs and CD4(+) T cells, and it may therefore act as a cell-intrinsic safeguard against inappropriate CTL differentiation and immunopathology.
Collapse
Affiliation(s)
- Shaun O'Brien
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Rajan M Thomas
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104; and
| | - Gerald B Wertheim
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104; and
| | - Fuqin Zhang
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Hao Shen
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Andrew D Wells
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104; and
| |
Collapse
|
32
|
Yin G, Qin M, Liu X, Suo J, Tang X, Tao G, Han Q, Suo X, Wu W. An Eimeria vaccine candidate based on Eimeria tenella immune mapped protein 1 and the TLR-5 agonist Salmonella typhimurium FliC flagellin. Biochem Biophys Res Commun 2013; 440:437-42. [PMID: 24076159 DOI: 10.1016/j.bbrc.2013.09.088] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 09/17/2013] [Indexed: 01/04/2023]
Abstract
Immune mapped protein-1 (IMP1) is a new protective protein in apicomplexan parasites, and exits in Eimeria tenella. But its structure and immunogenicity in E. tenella are still unknown. In this study, IMPI in E. tenella was predicted to be a membrane protein. To evaluate immunogenicity of IMPI in E. tenella, a chimeric subunit vaccine consisting of E. tenella IMP1 (EtIMP1) and a molecular adjuvant (a truncated flagellin, FliC) was constructed and over-expressed in Escherichia coli and its efficacy against E. tenella infection was evaluated. Three-week-old AA broiler chickens were vaccinated with the recombinant EtIMP1-truncated FliC without adjuvant or EtIMP1 with Freund's Complete Adjuvant. Immunization of chickens with the recombinant EtIMP1-truncated FliC fusion protein resulted in stronger cellular immune responses than immunization with only recombinant EtIMP1 with adjuvant. The clinical effect of the EtIMP1-truncated FliC without adjuvant was also greater than that of the EtIMP1 with adjuvant, which was evidenced by the differences between the two groups in body weight gain, oocyst output and caecal lesions of E. tenella-challenged chickens. The results suggested that the EtIMP1-flagellin fusion protein can be used as an effective immunogen in the development of subunit vaccines against Eimeria infection. This is the first demonstration of antigen-specific protective immunity against avian coccidiosis using a recombinant flagellin as an apicomplexan parasite vaccine adjuvant in chickens.
Collapse
Affiliation(s)
- Guangwen Yin
- National Animal Protozoa Laboratory and College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Jensen S, Steffensen MA, Jensen BAH, Schlüter D, Christensen JP, Thomsen AR. Adenovirus-Based Vaccine againstListeria monocytogenes: Extending the Concept of Invariant Chain Linkage. THE JOURNAL OF IMMUNOLOGY 2013; 191:4152-64. [DOI: 10.4049/jimmunol.1301290] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
34
|
Recombinant Salmonella enterica serovar Typhimurium as a vaccine vector for HIV-1 Gag. Viruses 2013; 5:2062-78. [PMID: 23989890 PMCID: PMC3798890 DOI: 10.3390/v5092062] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2013] [Revised: 08/05/2013] [Accepted: 08/22/2013] [Indexed: 01/30/2023] Open
Abstract
The HIV/AIDS epidemic remains a global health problem, especially in Sub-Saharan Africa. An effective HIV-1 vaccine is therefore badly required to mitigate this ever-expanding problem. Since HIV-1 infects its host through the mucosal surface, a vaccine for the virus needs to trigger mucosal as well as systemic immune responses. Oral, attenuated recombinant Salmonella vaccines offer this potential of delivering HIV-1 antigens to both the mucosal and systemic compartments of the immune system. So far, a number of pre-clinical studies have been performed, in which HIV-1 Gag, a highly conserved viral antigen possessing both T- and B-cell epitopes, was successfully delivered by recombinant Salmonella vaccines and, in most cases, induced HIV-specific immune responses. In this review, the potential use of Salmonella enterica serovar Typhimurium as a live vaccine vector for HIV-1 Gag is explored.
Collapse
|
35
|
Liu D, Ferrer IR, Konomos M, Ford ML. Inhibition of CD8+ T cell-derived CD40 signals is necessary but not sufficient for Foxp3+ induced regulatory T cell generation in vivo. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 191:1957-64. [PMID: 23858029 PMCID: PMC3735715 DOI: 10.4049/jimmunol.1300267] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Current models of CD4(+) T cell help suggest a major role for CD154 binding to CD40 expressed on dendritic cells, with a lesser role for direct T:T interactions via CD40 expressed on CD8(+) T cells. However, the contribution of CD8(+) T cell-derived CD40 signals during the donor-reactive T cell response to a transplant has never been studied. In this study, we examined the graft-rejection kinetics and CD4(+) and CD8(+) donor-reactive T cell responses under conditions in which CD40 was genetically ablated only on APC, as well as under conditions in which CD40 was genetically ablated only on donor-reactive CD8(+) T cells. Our results revealed a significant role for CD8(+) T cell-expressed CD40 in the augmentation of donor-reactive CD8(+) T cell responses following transplantation and showed that CD40 expressed on CD8(+) T cells must be inhibited to allow conversion of CD4(+) T cells into induced regulatory T cells. Thus, this study identifies a major role for CD8(+) T cell-derived CD40 signals as a critical switch factor that both promotes optimal differentiation of cytokine-producing CD8(+) effector T cell responses and inhibits the differentiation of Ag-specific Foxp3(+) induced regulatory T cells in vivo.
Collapse
Affiliation(s)
- Danya Liu
- Emory Transplant Center and Department of Surgery, Atlanta GA 30322
| | - Ivana R. Ferrer
- Emory Transplant Center and Department of Surgery, Atlanta GA 30322
| | - Michael Konomos
- Emory Transplant Center and Department of Surgery, Atlanta GA 30322
| | - Mandy L. Ford
- Emory Transplant Center and Department of Surgery, Atlanta GA 30322
| |
Collapse
|
36
|
Grubaugh D, Flechtner JB, Higgins DE. Proteins as T cell antigens: Methods for high-throughput identification. Vaccine 2013; 31:3805-10. [DOI: 10.1016/j.vaccine.2013.06.046] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 06/03/2013] [Accepted: 06/13/2013] [Indexed: 12/22/2022]
|
37
|
Yin G, Qin M, Liu X, Suo J, Suo X. Expression of Toxoplasma gondii dense granule protein7 (GRA7) in Eimeria tenella. Parasitol Res 2013; 112:2105-9. [DOI: 10.1007/s00436-013-3307-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2012] [Accepted: 01/18/2013] [Indexed: 10/27/2022]
|
38
|
Barat S, Willer Y, Rizos K, Claudi B, Mazé A, Schemmer AK, Kirchhoff D, Schmidt A, Burton N, Bumann D. Immunity to intracellular Salmonella depends on surface-associated antigens. PLoS Pathog 2012; 8:e1002966. [PMID: 23093937 PMCID: PMC3475680 DOI: 10.1371/journal.ppat.1002966] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 08/14/2012] [Indexed: 01/05/2023] Open
Abstract
Invasive Salmonella infection is an important health problem that is worsening because of rising antimicrobial resistance and changing Salmonella serovar spectrum. Novel vaccines with broad serovar coverage are needed, but suitable protective antigens remain largely unknown. Here, we tested 37 broadly conserved Salmonella antigens in a mouse typhoid fever model, and identified antigen candidates that conferred partial protection against lethal disease. Antigen properties such as high in vivo abundance or immunodominance in convalescent individuals were not required for protectivity, but all promising antigen candidates were associated with the Salmonella surface. Surprisingly, this was not due to superior immunogenicity of surface antigens compared to internal antigens as had been suggested by previous studies and novel findings for CD4 T cell responses to model antigens. Confocal microscopy of infected tissues revealed that many live Salmonella resided alone in infected host macrophages with no damaged Salmonella releasing internal antigens in their vicinity. In the absence of accessible internal antigens, detection of these infected cells might require CD4 T cell recognition of Salmonella surface-associated antigens that could be processed and presented even from intact Salmonella. In conclusion, our findings might pave the way for development of an efficacious Salmonella vaccine with broad serovar coverage, and suggest a similar crucial role of surface antigens for immunity to both extracellular and intracellular pathogens. Salmonella infections cause extensive morbidity and mortality worldwide. A vaccine that prevents systemic Salmonella infections is urgently needed but suitable antigens remain largely unknown. In this study we identified several antigen candidates that mediated protective immunity to Salmonella in a mouse typhoid fever model. Interestingly, all these antigens were associated with the Salmonella surface. This suggested that similar antigen properties might be relevant for CD4 T cell dependent immunity to intracellular pathogens like Salmonella, as for antibody-dependent immunity to extracellular pathogens. Detailed analysis revealed that Salmonella surface antigens were not generally more immunogenic compared to internal antigens. However, internal antigens were inaccessible for CD4 T cell recognition of a substantial number of infected host cells that contained exclusively live intact Salmonella. Together, these results might pave the way for development of an efficacious Salmonella vaccine, and provide a basis to facilitate antigen identification for Salmonella and possibly other intracellular pathogens.
Collapse
Affiliation(s)
- Somedutta Barat
- Focal Area Infection Biology, Biozentrum, University of Basel, Basel, Switzerland
| | - Yvonne Willer
- Junior Group “Mucosal Infections”, Hannover Medical School, Hannover, Germany
| | - Konstantin Rizos
- Department of Molecular Biology, Max-Planck-Institute for Infection Biology, Berlin, Germany
| | - Beatrice Claudi
- Focal Area Infection Biology, Biozentrum, University of Basel, Basel, Switzerland
| | - Alain Mazé
- Focal Area Infection Biology, Biozentrum, University of Basel, Basel, Switzerland
| | - Anne K. Schemmer
- Focal Area Infection Biology, Biozentrum, University of Basel, Basel, Switzerland
| | - Dennis Kirchhoff
- Immunomodulation Group, Deutsches Rheuma-Forschungszentrum Berlin, Berlin, Germany
| | - Alexander Schmidt
- Proteomics Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Neil Burton
- Focal Area Infection Biology, Biozentrum, University of Basel, Basel, Switzerland
| | - Dirk Bumann
- Focal Area Infection Biology, Biozentrum, University of Basel, Basel, Switzerland
- Junior Group “Mucosal Infections”, Hannover Medical School, Hannover, Germany
- Department of Molecular Biology, Max-Planck-Institute for Infection Biology, Berlin, Germany
- * E-mail:
| |
Collapse
|
39
|
Pham NLL, Badovinac VP, Harty JT. Epitope specificity of memory CD8+ T cells dictates vaccination-induced mortality in LCMV-infected perforin-deficient mice. Eur J Immunol 2012; 42:1488-99. [PMID: 22678903 PMCID: PMC3650624 DOI: 10.1002/eji.201142263] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Perforin-deficient (PKO) mice serve as models for familial hemophagocytic lympho-histiocytosis, a uniformly fatal disease associated with viral infection of perforin-deficient humans. Naïve perforin-deficient BALB/c mice survive while vaccinated PKO mice containing virus-specific memory CD8(+) T cells rapidly succumb to lymphocytic choriomeningitis virus (LCMV) infection. Thus, vaccination converts a nonlethal persistent infection into a fatal disease mediated by virus-specific memory CD8(+) T cells. Here, we determine the extent to which vaccination-induced mortality in PKO mice following LCMV challenge is due to differences in vaccine modalities, the quantity or epitope specificity of memory CD8(+) T cells. We show that LCMV-induced mortality in immune PKO mice is independent of vaccine modalities and that the starting number of memory CD8(+) T cells specific to the immunodominant epitope NP(118-126) dictates the magnitude of secondary CD8(+) T-cell expansion, the inability to regulate production of CD8(+) T-cell-derived IFN-γ, and mortality in the vaccinated PKO mice. Importantly, mortality is determined by the epitope specificity of memory CD8(+) T cells and the associated degree of functional exhaustion and cytokine dysregulation but not the absolute magnitude of CD8(+) T-cell expansion. These data suggest that deeper understanding of the parameters that influence the outcome of vaccine-induced diseases would aid rational vaccine design to minimize adverse outcomes after infection.
Collapse
Affiliation(s)
- Nhat-Long L. Pham
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa 52242, USA
| | - Vladimir P. Badovinac
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa 52242, USA
- Department of Pathology, University of Iowa, Iowa City, Iowa 52242, USA
| | - John T. Harty
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa 52242, USA
- Department of Pathology, University of Iowa, Iowa City, Iowa 52242, USA
- Department of Microbiology, University of Iowa, Iowa City, Iowa 52242, USA
| |
Collapse
|
40
|
Abstract
CD8 T cells play a critical role in the control and eradication of intracellular pathogens. Increased understanding of CD8 T cell biology provides insight that can be translated into improved vaccination strategies. The intracellular bacterium, Listeria monocytogenes, has been used as a model organism to study every phase of the CD8 T cell response to intracellular bacterial infection. Infection of laboratory mice with L. monocytogenes has provided insight into the factors that are involved in primary T cell responses, memory CD8 T cell generation, maintenance, functionality, and diversification following repeated pathogenic challenges. In this review, we will focus on work from our laboratories utilizing the murine model of L. monocytogenes to investigate the characteristics of CD8 T cell responses to infection. This model has profoundly advanced our understanding of the CD8 T cell response to infection and is likely to continue to provide invaluable basic insights that can be translated into the development of effective vaccination strategies to protect against pathogens.
Collapse
|
41
|
Zhu J, Larson CB, Ramaker MA, Quandt K, Wendte JM, Ku KP, Chen F, Jourdian GW, Vemulapalli R, Schurig GG, He Y. Characterization of recombinant B. abortus strain RB51SOD toward understanding the uncorrelated innate and adaptive immune responses induced by RB51SOD compared to its parent vaccine strain RB51. Front Cell Infect Microbiol 2011; 1:10. [PMID: 22919576 PMCID: PMC3417361 DOI: 10.3389/fcimb.2011.00010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 11/09/2011] [Indexed: 01/18/2023] Open
Abstract
Brucella abortus is a Gram-negative, facultative intracellular pathogen for several mammals, including humans. Live attenuated B. abortus strain RB51 is currently the official vaccine used against bovine brucellosis in the United States and several other countries. Overexpression of protective B. abortus antigen Cu/Zn superoxide dismutase (SOD) in a recombinant strain of RB51 (strain RB51SOD) significantly increases its vaccine efficacy against virulent B. abortus challenge in a mouse model. An attempt has been made to better understand the mechanism of the enhanced protective immunity of RB51SOD compared to its parent strain RB51. We previously reported that RB51SOD stimulated enhanced Th1 immune response. In this study, we further found that T effector cells derived from RB51SOD-immunized mice exhibited significantly higher cytotoxic T lymphocyte activity than T effector cells derived from RB51-immunized mice against virulent B. abortus-infected target cells. Meanwhile, the macrophage responses to these two strains were also studied. Compared to RB51, RB51SOD cells had a lower survival rate in macrophages and induced lower levels of macrophage apoptosis and necrosis. The decreased survival of RB51SOD cells correlates with the higher sensitivity of RB51SOD, compared to RB51, to the bactericidal action of either Polymyxin B or sodium dodecyl sulfate (SDS). Furthermore, a physical damage to the outer membrane of RB51SOD was observed by electron microscopy. Possibly due to the physical damage, overexpressed Cu/Zn SOD in RB51SOD was found to be released into the bacterial cell culture medium. Therefore, the stronger adaptive immunity induced by RB51SOD did not correlate with the low level of innate immunity induced by RB51SOD compared to RB51. This unique and apparently contradictory profile is likely associated with the differences in outer membrane integrity and Cu/Zn SOD release.
Collapse
MESH Headings
- Adaptive Immunity
- Animals
- Apoptosis
- Bacterial Proteins/genetics
- Brucella Vaccine/genetics
- Brucella Vaccine/immunology
- Brucella abortus/enzymology
- Brucella abortus/genetics
- Brucella abortus/immunology
- Brucella abortus/pathogenicity
- Brucellosis/immunology
- Brucellosis/prevention & control
- Cattle
- Cell Membrane/ultrastructure
- Detergents/pharmacology
- Disease Models, Animal
- Drug Resistance, Bacterial
- Humans
- Immunity, Innate
- Macrophages/immunology
- Macrophages/microbiology
- Mice
- Microscopy, Electron, Transmission
- Polymyxin B/pharmacology
- Recombination, Genetic
- Superoxide Dismutase/genetics
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/microbiology
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- Jianguo Zhu
- Unit for Laboratory Animal Medicine, Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI, USA
- School of Agriculture and Biology, Shanghai Jiaotong UniversityShanghai, China
| | - Charles B. Larson
- Unit for Laboratory Animal Medicine, Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | - Megan Ann Ramaker
- School of Veterinary Medicine, University of Wisconsin-MadisonMadison, WI, USA
| | - Kimberly Quandt
- College of Veterinary Medicine, Michigan State UniversityEast Lansing, MI, USA
| | - Jered M. Wendte
- Center for Veterinary Health Sciences, Oklahoma State UniversityStillwater, OK, USA
| | - Kimberly P. Ku
- Unit for Laboratory Animal Medicine, Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | - Fang Chen
- Unit for Laboratory Animal Medicine, Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | - George W. Jourdian
- Department of Internal Medicine and Department of Biological Chemistry, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | - Ramesh Vemulapalli
- Department of Comparative Pathobiology, School of Veterinary Medicine, Purdue UniversityWest Lafayette, IN, USA
| | - Gerhardt G. Schurig
- Center for Molecular Medicine and Infectious Diseases, Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Polytechnic Institute and State UniversityBlacksburg, VA, USA
| | - Yongqun He
- Unit for Laboratory Animal Medicine, Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI, USA
| |
Collapse
|
42
|
A rescue gone wrong. Nat Immunol 2011; 12:1137-8. [PMID: 22089212 DOI: 10.1038/ni.2161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
43
|
Huang X, Zou J, Xu H, Ding Y, Yin G, Liu X, Suo X. Transgenic Eimeria tenella Expressing Enhanced Yellow Fluorescent Protein Targeted to Different Cellular Compartments Stimulated Dichotomic Immune Responses in Chickens. THE JOURNAL OF IMMUNOLOGY 2011; 187:3595-602. [DOI: 10.4049/jimmunol.1100043] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
44
|
Zou J, Huang XX, Yin GW, Ding Y, Liu XY, Wang H, Chen QJ, Suo X. Evaluation of Toxoplasma gondii as a live vaccine vector in susceptible and resistant hosts. Parasit Vectors 2011; 4:168. [PMID: 21871123 PMCID: PMC3177786 DOI: 10.1186/1756-3305-4-168] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Accepted: 08/28/2011] [Indexed: 11/10/2022] Open
Abstract
Background Toxoplasma gondii has been shown to trigger strong cellular immune responses to heterologous antigens expressed by the parasite in the inbred mouse model [1]. We studied the immune response induced by T. gondii as an effective vaccine vector in chickens and rabbits. Results T. gondii RH strain was engineered to express the yellow fluorescent protein (YFP) in the cytoplasm. A subcutaneous injection of the transgenic T. gondii YFP in chickens afforded partial protection against the infection of transgenic E. tenella YFP. T. gondii YFP induced low levels of antibodies to YFP in chickens, suggesting that YFP specific cellular immune response was probably responsible for the protective immunity against E. tenella YFP infection. The measurement of T-cell response and IFN-γ production further confirmed that YFP specific Th1 mediated immune response was induced by T. gondii YFP in immunized chickens. The transgenic T. gondii stimulated significantly higher YFP specific IgG titers in rabbits than in chickens, suggesting greater immunogenicity in a T. gondii susceptible species than in a resistant species. Priming with T. gondii YFP and boosting with the recombinant YFP can induce a strong anti-YFP antibody response in both animal species. Conclusions Our findings suggest that T. gondii can be used as an effective vaccine vector and future research should focus on exploring avirulent no cyst-forming strains of T. gondii as a live vaccine vector in animals.
Collapse
Affiliation(s)
- Jun Zou
- National Animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Gregg B, Dzierszinski F, Tait E, Jordan KA, Hunter CA, Roos DS. Subcellular antigen location influences T-cell activation during acute infection with Toxoplasma gondii. PLoS One 2011; 6:e22936. [PMID: 21829561 PMCID: PMC3145783 DOI: 10.1371/journal.pone.0022936] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Accepted: 07/04/2011] [Indexed: 11/19/2022] Open
Abstract
Effective control of the intracellular protozoan parasite Toxoplasma gondii depends on the activation of antigen-specific CD8(+) T-cells that manage acute disease and prevent recrudescence during chronic infection. T-cell activation in turn, requires presentation of parasite antigens by MHC-I molecules on the surface of antigen presenting cells. CD8(+) T-cell epitopes have been defined for several T. gondii proteins, but it is unclear how these antigens enter into the presentation pathway. We have exploited the well-characterized model antigen ovalbumin (OVA) to investigate the ability of parasite proteins to enter the MHC-I presentation pathway, by engineering recombinant expression in various organelles. CD8(+) T-cell activation was assayed using 'B3Z' reporter cells in vitro, or adoptively-transferred OVA-specific 'OT-I' CD8(+) T-cells in vivo. As expected, OVA secreted into the parasitophorous vacuole strongly stimulated antigen-presenting cells. Lower levels of activation were observed using glycophosphatidyl inositol (GPI) anchored OVA associated with (or shed from) the parasite surface. Little CD8(+) T-cell activation was detected using parasites expressing intracellular OVA in the cytosol, mitochondrion, or inner membrane complex (IMC). These results indicate that effective presentation of parasite proteins to CD8(+) T-cells is a consequence of active protein secretion by T. gondii and escape from the parasitophorous vacuole, rather than degradation of phagocytosed parasites or parasite products.
Collapse
Affiliation(s)
- Beth Gregg
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Florence Dzierszinski
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Elia Tait
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Kimberly A. Jordan
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Christopher A. Hunter
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - David S. Roos
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
46
|
Yin G, Liu X, Zou J, Huang X, Suo X. Co-expression of reporter genes in the widespread pathogen Eimeria tenella using a double-cassette expression vector strategy. Int J Parasitol 2011; 41:813-6. [PMID: 21550346 DOI: 10.1016/j.ijpara.2011.04.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2011] [Revised: 03/31/2011] [Accepted: 04/04/2011] [Indexed: 11/26/2022]
Abstract
The double-cassette expression vector strategy is valuable for many studies, including comparative analysis of the function of promoters and expression of genes in different compartments. In this study, we report co-expression of enhanced yellow fluorescent protein (EYFP) and red fluorescent protein (RFP) in Eimeria tenella transfected with two double-cassette expression vectors, pMIC-EYFP/ACT-RFP and pMIC-EYFP/ACTss-RFP. The results showed that under regulation of the mic1 promoter, EYFP was expressed in sporulated oocysts but not in unsporulated ones, while under regulation of the actin promoter RFP was expressed in both forms. We found that the signal peptide of Toxoplasma gondii dense granule protein 8 (GRA8) located the RFP expression to the parasitophorous vacuoles of the parasites, the margins of the unsporulated oocysts and the cavities of the sporocysts. The feasibility of co-expression of exogenous proteins in E. tenella is important for the development of transgenic E. tenella as a novel vaccine vector.
Collapse
Affiliation(s)
- Guangwen Yin
- National Animal Protozoa Laboratory & College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | | | | | | | | |
Collapse
|
47
|
Evaluation of oral immunization with recombinant avian influenza virus HA1 displayed on the Lactococcus lactis surface and combined with the mucosal adjuvant cholera toxin subunit B. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2011; 18:1046-51. [PMID: 21632890 DOI: 10.1128/cvi.00050-11] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The development of safe and efficient avian influenza vaccines for human and animal uses is essential for preventing virulent outbreaks and pandemics worldwide. In this study, we constructed a recombinant (pgsA-HA1 gene fusion) Lactococcus lactis strain that expresses and displays the avian influenza virus HA1 antigens on its surface. The vectors were administered by oral delivery with or without the addition of cholera toxin subunit B (CTB). The resulting immune responses were analyzed, and the mice were eventually challenged with lethal doses of H5N1 viruses. Significant titers of hemagglutinin (HA)-specific serum IgG and fecal IgA were detected in the group that also received CTB. Cellular immunities were also shown in both cell proliferation and gamma interferon (IFN-γ) enzyme-linked immunospot (ELISpot) assays. Most importantly, the mice that received the L. lactis pgsA-HA1 strain combined with CTB were completely protected from lethal challenge of the H5N1 virus. These findings support the further development of L. lactis-based avian influenza virus vaccines for human and animal uses.
Collapse
|
48
|
Campisi L, Soudja SM, Cazareth J, Bassand D, Lazzari A, Brau F, Narni-Mancinelli E, Glaichenhaus N, Geissmann F, Lauvau G. Splenic CD8α⁺ dendritic cells undergo rapid programming by cytosolic bacteria and inflammation to induce protective CD8⁺ T-cell memory. Eur J Immunol 2011; 41:1594-605. [PMID: 21469106 DOI: 10.1002/eji.201041036] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Revised: 01/08/2011] [Accepted: 03/09/2011] [Indexed: 11/09/2022]
Abstract
Memory CD8(+) T lymphocytes are critical effector cells of the adaptive immune system mediating long-lived pathogen-specific protective immunity. Three signals - antigen, costimulation and inflammation - orchestrate optimal CD8(+) T-cell priming and differentiation into effector and memory cells and shape T-cell functional fate and ability to protect against challenge infections. While among the conventional spleen DCs (cDCs), the CD8α(+) but not the CD8α(-) cDCs most efficiently mediate CD8(+) T-cell priming, it is unclear which subset, irrespective of their capacity to process MHC class I-associated antigens, is most efficient at inducing naïve CD8(+) T-cell differentiation into pathogen-specific protective memory cells in vivo. Moreover, the origin of the required signals is still unclear. Using mice infected with the intracellular bacterium Listeria monocytogenes, we show that splenic CD8α(+) cDCs become endowed with all functional features to optimally prime protective memory CD8(+) T cells in vivo within only a few hours post-immunization. Such programming requires both cytosolic signals resulting from bacterial invasion of the host cells and extracellular inflammatory mediators. Thus, these data designate these cells as the best candidates to facilitate the development of cell-based vaccine therapy.
Collapse
Affiliation(s)
- Laura Campisi
- Institut National de Santé et de Recherche Médicale Unité 924, Groupe Avenir, Valbonne, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Nolz JC, Harty JT. Protective capacity of memory CD8+ T cells is dictated by antigen exposure history and nature of the infection. Immunity 2011; 34:781-93. [PMID: 21549619 DOI: 10.1016/j.immuni.2011.03.020] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 12/09/2010] [Accepted: 03/08/2011] [Indexed: 12/12/2022]
Abstract
Infection or vaccination confers heightened resistance to pathogen rechallenge because of quantitative and qualitative differences between naive and primary memory T cells. Herein, we show that secondary (boosted) memory CD8+ T cells were better than primary memory CD8+ T cells in controlling some, but not all acute infections with diverse pathogens. However, secondary memory CD8+ T cells were less efficient than an equal number of primary memory cells at preventing chronic LCMV infection and are more susceptible to functional exhaustion. Importantly, localization of memory CD8+ T cells within lymph nodes, which is reduced by antigen restimulation, was critical for both viral control in lymph nodes and for the sustained CD8+ T cell response required to prevent chronic LCMV infection. Thus, repeated antigen stimulation shapes memory CD8+ T cell populations to either enhance or decrease per cell protective immunity in a pathogen-specific manner, a concept of importance in vaccine design against specific diseases.
Collapse
Affiliation(s)
- Jeffrey C Nolz
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA
| | | |
Collapse
|
50
|
Floyd TL, Koehn BH, Kitchens WH, Robertson JM, Cheeseman JA, Stempora L, Larsen CP, Ford ML. Limiting the amount and duration of antigen exposure during priming increases memory T cell requirement for costimulation during recall. THE JOURNAL OF IMMUNOLOGY 2011; 186:2033-41. [PMID: 21257960 DOI: 10.4049/jimmunol.1003015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Donor-reactive memory T cells (Tmem) can play an important role in mediating graft rejection after transplantation. Transplant recipients acquire donor-reactive Tmem not only through prior sensitization with alloantigens but also through previous exposure to environmental pathogens that are cross-reactive with allogeneic peptide-MHC complexes. Current dogma suggests that most, if not all, Tmem responses are independent of the requirement for CD28 and/or CD154/CD40-mediated costimulation to mount a recall response. However, heterogeneity among Tmem is increasingly being appreciated, and one important factor known to impact the function and phenotype of Ag-specific T cell responses is the amount/duration of Ag exposure. Importantly, the impact of Ag exposure on development of costimulation independence is currently unknown. In this study, we interrogated the effect of decreased Ag amount/duration during priming on the ability of donor-reactive Tmem to mediate costimulation blockade-resistant rejection during a recall response after transplantation in a murine model. Recipients possessing donor-reactive Tmem responses that were generated under conditions of reduced Ag exposure exhibited similar frequencies of Ag-specific T cells at day 30 postinfection, but, strikingly, failed to mediate costimulation blockade-resistant rejection after challenge with an OVA-expressing skin graft. Thus, these data demonstrate the amount/duration of Ag exposure is a critical factor in determining Tmem's relative requirement for costimulation during the recall response after transplantation.
Collapse
Affiliation(s)
- Tamara L Floyd
- Department of Surgery and Emory Transplant Center, Emory University, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | |
Collapse
|