1
|
Arnold F, Kupferschmid L, Weissenborn P, Heldmann L, Hummel JF, Zareba P, Sagar, Rogg M, Schell C, Tanriver Y. Tissue-resident memory T cells break tolerance to renal autoantigens and orchestrate immune-mediated nephritis. Cell Mol Immunol 2024; 21:1066-1081. [PMID: 38961265 PMCID: PMC11364874 DOI: 10.1038/s41423-024-01197-z] [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: 02/08/2024] [Accepted: 06/13/2024] [Indexed: 07/05/2024] Open
Abstract
Immune-mediated nephritis is a leading cause of acute kidney injury and chronic kidney disease. While the role of B cells and antibodies has been extensively investigated in the past, the advent of immune-checkpoint inhibitors has led to a reappraisal of the role of T cells in renal immunology. However, it remains elusive how T cells with specificity for renal autoantigens are activated and participate in immune-mediated nephritis. Here, we followed the fate and function of pathogen-activated autoreactive CD8 T cells that are specific for a renal autoantigen. We demonstrate that recently activated splenic CD8 T cells developed a hybrid phenotype in the context of renal autoantigen cross-presentation, combining hallmarks of activation and T cell dysfunction. While circulating memory T cells rapidly disappeared, tissue-resident memory T cells emerged and persisted within the kidney, orchestrating immune-mediated nephritis. Notably, T cells infiltrating kidneys of patients with interstitial nephritis also expressed key markers of tissue residency. This study unveils how a tissue-specific immune response can dissociate from its systemic counterpart driving a compartmentalized immune response in the kidneys of mice and man. Consequently, targeting tissue-resident memory T cells emerges as a promising strategy to control immune-mediated kidney disease.
Collapse
Affiliation(s)
- Frederic Arnold
- Department of Medicine IV, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Institute of Microbiology and Hygiene, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Laurence Kupferschmid
- Institute of Microbiology and Hygiene, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Philipp Weissenborn
- Institute of Microbiology and Hygiene, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lukas Heldmann
- Institute of Microbiology and Hygiene, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jonas F Hummel
- Institute of Microbiology and Hygiene, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Paulina Zareba
- Institute of Pathology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sagar
- Department of Medicine II, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Manuel Rogg
- Institute of Pathology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christoph Schell
- Institute of Pathology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Yakup Tanriver
- Department of Medicine IV, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute of Microbiology and Hygiene, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
2
|
Sunga GM, Hartgerink J, Sikora AG, Young S. Enhancement of Immunotherapies in Head and Neck Cancers Using Biomaterial-Based Treatment Strategies. Tissue Eng Part C Methods 2023; 29:257-275. [PMID: 37183412 PMCID: PMC10282827 DOI: 10.1089/ten.tec.2023.0090] [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: 04/24/2023] [Accepted: 05/12/2023] [Indexed: 05/16/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a challenging disease to treat because of typically late-stage diagnoses and tumor formation in difficult-to-treat areas, sensitive to aggressive or invasive treatments. To date, HNSCC treatments have been limited to surgery, radiotherapy, and chemotherapy, which may have significant morbidity and often lead to long-lasting side effects. The development of immunotherapies has revolutionized cancer treatment by providing a promising alternative to standard-of-care therapies. However, single-agent immunotherapy has been only modestly effective in the treatment of various cancers, including HNSCC, with most patients receiving no overall benefit or increased survival. In addition, single-agent immunotherapy's limitations, namely immune-related side effects and the necessity of multidose treatments, must be addressed to further improve treatment efficacy. Biocompatible biomaterials, in combination with cancer immunotherapies, offer numerous advantages in the concentration, localization, and controlled release of drugs, cancer antigens, and immune cells. Biomaterial structures are diverse, and their design can generally be customized to enhance immunotherapy response. In preclinical settings, the use of biomaterials has shown great promise in improving the efficacy of single-agent immunotherapy. Herein, we provide an overview of current immunotherapy treatments for HNSCC and their limitations, as well as the potential applications of biomaterials in enhancing cancer immunotherapies. Impact Statement Advances in anticancer immunotherapies for the past 30 years have yielded exciting clinical results and provided alternatives to long-standing standard-of-care treatments, which are associated with significant toxicities and long-term morbidity. However, patients with head and neck squamous cell carcinoma (HNSCC) have not benefited from immunotherapies as much as patients with other cancers. Immunotherapy limitations include systemic side effects, therapeutic resistance, poor delivery kinetics, and limited patient responses. Biomaterial-enhanced immunotherapies, as explored in this review, are a potentially powerful means of achieving localized drug delivery, sustained and controlled drug release, and immunomodulation. They may overcome current treatment limitations and improve patient outcomes and care.
Collapse
Affiliation(s)
- Gemalene M. Sunga
- Katz Department of Oral and Maxillofacial Surgery, The University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
| | - Jeffrey Hartgerink
- Department of Chemistry, Rice University, Houston, Texas, USA
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Andrew G. Sikora
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Simon Young
- Katz Department of Oral and Maxillofacial Surgery, The University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
| |
Collapse
|
3
|
Ma Y, Sannino D, Linden JR, Haigh S, Zhao B, Grigg JB, Zumbo P, Dündar F, Butler D, Profaci CP, Telesford K, Winokur PN, Rumah KR, Gauthier SA, Fischetti VA, McClane BA, Uzal FA, Zexter L, Mazzucco M, Rudick R, Danko D, Balmuth E, Nealon N, Perumal J, Kaunzner U, Brito IL, Chen Z, Xiang JZ, Betel D, Daneman R, Sonnenberg GF, Mason CE, Vartanian T. Epsilon toxin-producing Clostridium perfringens colonize the multiple sclerosis gut microbiome overcoming CNS immune privilege. J Clin Invest 2023; 133:e163239. [PMID: 36853799 PMCID: PMC10145940 DOI: 10.1172/jci163239] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 02/23/2023] [Indexed: 03/01/2023] Open
Abstract
Multiple sclerosis (MS) is a complex disease of the CNS thought to require an environmental trigger. Gut dysbiosis is common in MS, but specific causative species are unknown. To address this knowledge gap, we used sensitive and quantitative PCR detection to show that people with MS were more likely to harbor and show a greater abundance of epsilon toxin-producing (ETX-producing) strains of C. perfringens within their gut microbiomes compared with individuals who are healthy controls (HCs). Isolates derived from patients with MS produced functional ETX and had a genetic architecture typical of highly conjugative plasmids. In the active immunization model of experimental autoimmune encephalomyelitis (EAE), where pertussis toxin (PTX) is used to overcome CNS immune privilege, ETX can substitute for PTX. In contrast to PTX-induced EAE, where inflammatory demyelination is largely restricted to the spinal cord, ETX-induced EAE caused demyelination in the corpus callosum, thalamus, cerebellum, brainstem, and spinal cord, more akin to the neuroanatomical lesion distribution seen in MS. CNS endothelial cell transcriptional profiles revealed ETX-induced genes that are known to play a role in overcoming CNS immune privilege. Together, these findings suggest that ETX-producing C. perfringens strains are biologically plausible pathogens in MS that trigger inflammatory demyelination in the context of circulating myelin autoreactive lymphocytes.
Collapse
Affiliation(s)
- Yinghua Ma
- Feil Family Brain and Mind Research Institute
| | | | | | | | - Baohua Zhao
- Feil Family Brain and Mind Research Institute
| | - John B. Grigg
- Jill Roberts Institute for Research in Inflammatory Bowel Disease
- Joan and Sanford I. Weill Department of Medicine, and
- Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, New York, USA
- Immunology and Microbial Pathogenesis Program and
| | - Paul Zumbo
- Applied Bioinformatics Core, Division of Hematology/Oncology, Department of Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, New York, USA
- Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell Medical College, Cornell University, New York, New York, USA
| | - Friederike Dündar
- Applied Bioinformatics Core, Division of Hematology/Oncology, Department of Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, New York, USA
- Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell Medical College, Cornell University, New York, New York, USA
| | - Daniel Butler
- Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell Medical College, Cornell University, New York, New York, USA
| | - Caterina P. Profaci
- Departments of Pharmacology and Neurosciences, UCSD, San Diego, California, USA
| | | | - Paige N. Winokur
- Harold and Margaret Milliken Hatch Laboratory of Neuro-endocrinology and
| | - Kareem R. Rumah
- Laboratory of Bacterial Pathogenesis and Immunology, Rockefeller University, New York, New York, USA
| | - Susan A. Gauthier
- Department of Neurology, Weill Cornell Medical College, Cornell University, New York, New York, USA
| | - Vincent A. Fischetti
- Laboratory of Bacterial Pathogenesis and Immunology, Rockefeller University, New York, New York, USA
| | - Bruce A. McClane
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Francisco A. Uzal
- California Animal Health and Food Safety Laboratory, School of Veterinary Medicine, UCD, Davis, California, USA
| | - Lily Zexter
- Department of Neurology, Weill Cornell Medical College, Cornell University, New York, New York, USA
| | | | | | - David Danko
- Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell Medical College, Cornell University, New York, New York, USA
| | | | - Nancy Nealon
- Department of Neurology, Weill Cornell Medical College, Cornell University, New York, New York, USA
| | - Jai Perumal
- Department of Neurology, Weill Cornell Medical College, Cornell University, New York, New York, USA
| | - Ulrike Kaunzner
- Department of Neurology, Weill Cornell Medical College, Cornell University, New York, New York, USA
| | - Ilana L. Brito
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, USA
| | - Zhengming Chen
- Division of Biostatistics, Department of Population Health Sciences, and
| | - Jenny Z. Xiang
- Genomics Resources Core Facility, Core Laboratories Center, Weill Cornell Medicine, New York, New York, USA
| | - Doron Betel
- Applied Bioinformatics Core, Division of Hematology/Oncology, Department of Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, New York, USA
- Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell Medical College, Cornell University, New York, New York, USA
| | - Richard Daneman
- Departments of Pharmacology and Neurosciences, UCSD, San Diego, California, USA
| | - Gregory F. Sonnenberg
- Jill Roberts Institute for Research in Inflammatory Bowel Disease
- Joan and Sanford I. Weill Department of Medicine, and
- Department of Microbiology and Immunology, Weill Cornell Medical College, Cornell University, New York, New York, USA
- Immunology and Microbial Pathogenesis Program and
| | - Christopher E. Mason
- Feil Family Brain and Mind Research Institute
- Physiology and Biophysics, Institute for Computational Biomedicine, Weill Cornell Medical College, Cornell University, New York, New York, USA
| | - Timothy Vartanian
- Feil Family Brain and Mind Research Institute
- Immunology and Microbial Pathogenesis Program and
- Department of Neurology, Weill Cornell Medical College, Cornell University, New York, New York, USA
| |
Collapse
|
4
|
Ebensen T, Arntz A, Schulze K, Hanefeld A, Guzmán CA, Scherließ R. Pulmonary Application of Novel Antigen-Loaded Chitosan Nano-Particles Co-Administered with the Mucosal Adjuvant C-Di-AMP Resulted in Enhanced Immune Stimulation and Dose Sparing Capacity. Pharmaceutics 2023; 15:pharmaceutics15041238. [PMID: 37111723 PMCID: PMC10145907 DOI: 10.3390/pharmaceutics15041238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/31/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
The most successful medical intervention for preventing infectious diseases is still vaccination. This effective strategy has resulted in decreased mortality and extended life expectancy. However, there is still a critical need for novel vaccination strategies and vaccines. Antigen cargo delivery by nanoparticle-based carriers could promote superior protection against constantly emerging viruses and subsequent diseases. This should be sustained by the induction of vigorous cellular and humoral immunity, capable of acting both at the systemic and mucosal levels. Induction of antigen-specific responses at the portal of entry of pathogens is considered an important scientific challenge. Chitosan, which is widely regarded as a biodegradable, biocompatible and non-toxic material for functionalized nanocarriers, as well as having adjuvant activity, enables antigen administration via less-invasive mucosal routes such as sublingual or pulmonic application route. In this proof of principle study, we evaluate the efficacy of chitosan nanocarriers loaded with the model antigen Ovalbumin (OVA) co-administrated with the STING agonist bis-(3',5')-cyclic dimeric adenosine monophosphate (c-di-AMP) given by pulmonary route. Here, BALB/c mice were immunized with four doses of the formulation that stimulates enhanced antigen-specific IgG titers in sera. In addition, this vaccine formulation also promotes a strong Th1/Th17 response characterized by high secretion of IFN-γ, IL-2 and IL-17, as well as induction of CD8+ T cells. Furthermore, the novel formulation exhibited strong dose-sparing capacity, enabling a 90% reduction of the antigen concentration. Altogether, our results suggest that chitosan nanocarriers, in combination with the mucosal adjuvant c-di-AMP, are a promising technology platform for the development of innovative mucosal vaccines against respiratory pathogens (e.g., Influenza or RSV) or for therapeutic vaccines.
Collapse
Affiliation(s)
- Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Inhoffenstr. 7, 38124 Braunschweig, Germany
| | - Andrea Arntz
- Department of Pharmaceutics and Biopharmaceutics, Kiel University, Grasweg 9a, 24118 Kiel, Germany
| | - Kai Schulze
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Inhoffenstr. 7, 38124 Braunschweig, Germany
| | - Andrea Hanefeld
- Merck KGaA, Frankfurter Straße 250, 64293 Darmstadt, Germany
| | - Carlos A Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research (HZI), Inhoffenstr. 7, 38124 Braunschweig, Germany
| | - Regina Scherließ
- Department of Pharmaceutics and Biopharmaceutics, Kiel University, Grasweg 9a, 24118 Kiel, Germany
| |
Collapse
|
5
|
Ruterbusch M, Pruner KB, Shehata L, Pepper M. In Vivo CD4 + T Cell Differentiation and Function: Revisiting the Th1/Th2 Paradigm. Annu Rev Immunol 2021; 38:705-725. [PMID: 32340571 DOI: 10.1146/annurev-immunol-103019-085803] [Citation(s) in RCA: 293] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The discovery of CD4+ T cell subset-defining master transcription factors and framing of the Th1/Th2 paradigm ignited the CD4+ T cell field. Advances in in vivo experimental systems, however, have revealed that more complex lineage-defining transcriptional networks direct CD4+ T cell differentiation in the lymphoid organs and tissues. This review focuses on the layers of fate decisions that inform CD4+ T cell differentiation in vivo. Cytokine production by antigen-presenting cells and other innate cells influences the CD4+ T cell effector program [e.g., T helper type 1 (Th1), Th2, Th17]. Signals downstream of the T cell receptor influence whether individual clones bearing hallmarks of this effector program become T follicular helper cells, supporting development of B cells expressing specific antibody isotypes, or T effector cells, which activate microbicidal innate cells in tissues. These bifurcated, parallel axes allow CD4+ T cells to augment their particular effector program and prevent disease.
Collapse
Affiliation(s)
- Mikel Ruterbusch
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington 98109, USA; ,
| | - Kurt B Pruner
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington 98109, USA; ,
| | - Laila Shehata
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington 98109, USA; ,
| | - Marion Pepper
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington 98109, USA; ,
| |
Collapse
|
6
|
Mausberg AK, Szepanowski F, Odoardi F, Flügel A, Kleinschnitz C, Stettner M, Kieseier BC. Trapped in the epineurium: early entry into the endoneurium is restricted to neuritogenic T cells in experimental autoimmune neuritis. J Neuroinflammation 2018; 15:217. [PMID: 30068351 PMCID: PMC6090976 DOI: 10.1186/s12974-018-1259-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 07/19/2018] [Indexed: 12/24/2022] Open
Abstract
Background Autoimmune polyneuropathies are acquired inflammatory disorders of the peripheral nervous system (PNS) characterized by inflammation, demyelination, and axonal degeneration. Although the pathogenesis has not been fully elucidated, T cells recognizing self-antigens are believed to initiate inflammation in a subgroup of patients. However, the route and time of T cell entry into the PNS have not yet been described in detail. In this study, we analyzed both kinetics as well as localization of retrovirally transfected green fluorescent protein (GFP)-expressing neuritogenic T lymphocytes in experimental autoimmune neuritis (EAN). Methods T lymphocytes obtained from rats following EAN induction by immunization with peripheral nerve protein peptide P255–78 were retrovirally engineered to express GFP. Non-specific T cells were negatively selected by in vitro restimulation, whereas GFP-expressing neuritogenic T cells (reactive to P255–78) were adoptively transferred into healthy rats (AT-EAN). Antigen-specific T cell tracking and localization was performed by flow cytometry and immunohistochemistry during the course of disease. Results After induction of autoimmune neuritis, P2-reactive T cells were detectable in the liver, spleen, lymph nodes, lung, peripheral blood, and the sciatic nerves with distinct kinetics. A significant number of GFP+ T cells appeared early in the lung with a peak at day four. In the peripheral nerves within the first days, GFP-negative T cells rapidly accumulated and exceeded the number of GFP-expressing cells, but did not enter the endoneurium. Very early after adoptive transfer, T cells are found in proximity to peripheral nerves and in the epineurium. However, only GFP-expressing neuritogenic T cells are able to enter the endoneurium from day five after transfer. Conclusions Our findings suggest that neuritogenic T cells invade the PNS early in the course of disease. However, neuritogenic T cells cross the blood-nerve barrier with a certain delay without preference to dorsal roots. Further understanding of the pathophysiological role of autoagressive T cells may help to improve therapeutic strategies in immune-mediated neuropathies.
Collapse
Affiliation(s)
- Anne K Mausberg
- Department of Neurology, Research Group for Clinical and Experimental Neuroimmunology, University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany.
| | - Fabian Szepanowski
- Department of Neurology, Research Group for Clinical and Experimental Neuroimmunology, University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Francesca Odoardi
- Department of Neuroimmunology, University Medical Centre, Goettingen, Germany
| | - Alexander Flügel
- Department of Neuroimmunology, University Medical Centre, Goettingen, Germany
| | - Christoph Kleinschnitz
- Department of Neurology, Research Group for Clinical and Experimental Neuroimmunology, University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Mark Stettner
- Department of Neurology, Research Group for Clinical and Experimental Neuroimmunology, University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Bernd C Kieseier
- Department of Neurology, Medical Faculty, Heinrich Heine University Duesseldorf, 40225, Duesseldorf, Germany
| |
Collapse
|
7
|
Determining T-cell specificity to understand and treat disease. Nat Biomed Eng 2017; 1:784-795. [DOI: 10.1038/s41551-017-0143-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 09/05/2017] [Indexed: 02/06/2023]
|
8
|
Byun SH, Lee JH, Jung NC, Choi HJ, Song JY, Seo HG, Choi J, Jung SY, Kang S, Choi YS, Chung JH, Lim DS. Rosiglitazone-mediated dendritic cells ameliorate collagen-induced arthritis in mice. Biochem Pharmacol 2016; 115:85-93. [PMID: 27208887 DOI: 10.1016/j.bcp.2016.05.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/17/2016] [Indexed: 10/21/2022]
Abstract
Rosiglitazone is a selective ligand for peroxisome proliferator-activated receptor-gamma (PPAR-γ), which serves diverse biological functions. A number of autoimmune disease models have been used to examine the anti-inflammatory and immunosuppressive effects of tolerogenic dendritic cells (tDCs). The aim of the present study was to investigate whether rosiglitazone-mediated DC (Rosi-DC) therapy suppressed arthritis in a collagen-induced arthritis (CIA) mouse model. Rosi-DCs were generated by treating immature DCs with TNF-α, type II collagen, and rosiglitazone. CIA mice then received subcutaneously (s.c.) two injections of Rosi-DCs. The severity of arthritis was then assessed histopathologically. The phenotypes of the DC and regulatory T (Treg) cell populations in CIA mice were determined by flow cytometry and the effect of Rosi-DCs on the secretion of autoimmunity-inducing cytokines was examined by ELISA. Rosi-DCs expressed lower levels of DC-related surface markers than mature DCs. Histopathological examination revealed that the degree of inflammation in the paws of Rosi-DC-treated mice was much lower than that in the paws of PBS-treated CIA mice. Taken together, these results clearly show that rosiglitazone-mediated DCs ameliorate CIA, most likely via the induction of antigen-specific Treg cells.
Collapse
Affiliation(s)
- Sei-Hee Byun
- Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400, Republic of Korea
| | - Jun-Ho Lee
- Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400, Republic of Korea; Pharos Vaccine Inc., 545 Dunchon-daero, Jungwon-gu, Seongnam, Gyeonggi-do 462-807, Republic of Korea
| | - Nam-Chul Jung
- Pharos Vaccine Inc., 545 Dunchon-daero, Jungwon-gu, Seongnam, Gyeonggi-do 462-807, Republic of Korea
| | - Hyun-Ji Choi
- Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400, Republic of Korea
| | - Jie-Young Song
- Department of Radiation Cancer Sciences, Korea Institute of Radiological and Medical Sciences, 215-4 Gongneung-dong, Nowon-gu, Seoul 139-706, Republic of Korea
| | - Han Geuk Seo
- Department of Animal Biotechnology, Konkuk University, 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Jinjung Choi
- Division of Rheumatology, Bundang CHA Medical Center, Yatap-dong, Bundang-gu, Seongnam-si, Gyeonggi-do 463-712, Republic of Korea
| | - Sang Youn Jung
- Division of Rheumatology, Bundang CHA Medical Center, Yatap-dong, Bundang-gu, Seongnam-si, Gyeonggi-do 463-712, Republic of Korea
| | - Sangjin Kang
- Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400, Republic of Korea
| | - Yong-Soo Choi
- Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400, Republic of Korea
| | - Ji Hyung Chung
- Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400, Republic of Korea
| | - Dae-Seog Lim
- Department of Biotechnology, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 463-400, Republic of Korea.
| |
Collapse
|
9
|
Yang H, Lee J, Seo JH, Oh KH, Cho YH, Yoo YC. Induction of Oral Tolerance by Gamma-Irradiated Ovalbumin Administration. Korean J Food Sci Anim Resour 2016; 36:14-8. [PMID: 27499658 PMCID: PMC4973942 DOI: 10.5851/kosfa.2016.36.1.14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Revised: 11/13/2015] [Accepted: 11/18/2015] [Indexed: 11/06/2022] Open
Affiliation(s)
- Hui Yang
- Department of Microbiology, Konyang University, Daejeon 35365, Korea
- These authors contributed equally to this work
| | - Junglim Lee
- Department of Microbiology, Konyang University, Daejeon 35365, Korea
- Myunggok Medical Research Institute of Konyang University, Daejeon 35365, Korea
- These authors contributed equally to this work
| | - Ji Hyun Seo
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Korea
| | - Kwang Hoon Oh
- Department of Physical Education, Konju National University, Gongju 32688, Korea
| | - Young Ho Cho
- Department of Pharmaceutics & Biotechnology, Konyang University, Daejeon 35365, Korea
| | - Yung Choon Yoo
- Department of Microbiology, Konyang University, Daejeon 35365, Korea
- Myunggok Medical Research Institute of Konyang University, Daejeon 35365, Korea
- Corresponding author: Yung Choon Yoo, Department of Microbiology, Konyang University, Daejeon 35365, Korea. Tel: +82-42-600-6495, Fax: +82-42-600-6495, E-mail:
| |
Collapse
|
10
|
Boianelli A, Pettini E, Prota G, Medaglini D, Vicino A. A Stochastic Model for CD4+ T Cell Proliferation and Dissemination Network in Primary Immune Response. PLoS One 2015; 10:e0135787. [PMID: 26301680 PMCID: PMC4547705 DOI: 10.1371/journal.pone.0135787] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 07/27/2015] [Indexed: 01/29/2023] Open
Abstract
The study of the initial phase of the adaptive immune response after first antigen encounter provides essential information on the magnitude and quality of the immune response. This phase is characterized by proliferation and dissemination of T cells in the lymphoid organs. Modeling and identifying the key features of this phenomenon may provide a useful tool for the analysis and prediction of the effects of immunization. This knowledge can be effectively exploited in vaccinology, where it is of interest to evaluate and compare the responses to different vaccine formulations. The objective of this paper is to construct a stochastic model based on branching process theory, for the dissemination network of antigen-specific CD4+ T cells. The devised model is validated on in vivo animal experimental data. The model presented has been applied to the vaccine immunization context making references to simple proliferation laws that take into account division, death and quiescence, but it can also be applied to any context where it is of interest to study the dynamic evolution of a population.
Collapse
Affiliation(s)
- Alessandro Boianelli
- Systems Medicine of Infectious Diseases group and Braunschweig Integrated Centre of Systems Biology, Department of Systems Immunology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124 Braunschweig, Germany
- * E-mail:
| | - Elena Pettini
- Laboratorio di Microbiologia Molecolare e Biotecnologie, Dipartimento di Biotecnologie Mediche, Università di Siena, Viale Bracci 1, 53100 Siena, Italy
| | - Gennaro Prota
- Laboratorio di Microbiologia Molecolare e Biotecnologie, Dipartimento di Biotecnologie Mediche, Università di Siena, Viale Bracci 1, 53100 Siena, Italy
| | - Donata Medaglini
- Laboratorio di Microbiologia Molecolare e Biotecnologie, Dipartimento di Biotecnologie Mediche, Università di Siena, Viale Bracci 1, 53100 Siena, Italy
| | - Antonio Vicino
- Dipartimento di Ingegneria dell’Informazione e Science Matematiche, Università di Siena, Via Roma 56, 53100 Siena, Italy
| |
Collapse
|
11
|
Chang TT, Spurlock SM, Candelario TLT, Grenon SM, Hughes-Fulford M. Spaceflight impairs antigen-specific tolerance induction in vivo and increases inflammatory cytokines. FASEB J 2015; 29:4122-32. [PMID: 26085131 DOI: 10.1096/fj.15-275073] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 06/08/2015] [Indexed: 01/07/2023]
Abstract
The health risks of a dysregulated immune response during spaceflight are important to understand as plans emerge for humans to embark on long-term space travel to Mars. In this first-of-its-kind study, we used adoptive transfer of T-cell receptor transgenic OT-II CD4 T cells to track an in vivo antigen-specific immune response that was induced during the course of spaceflight. Experimental mice destined for spaceflight and mice that remained on the ground received transferred OT-II cells and cognate peptide stimulation with ovalbumin (OVA) 323-339 plus the inflammatory adjuvant, monophosphoryl lipid A. Control mice in both flight and ground cohorts received monophosphoryl lipid A alone without additional OVA stimulation. Numbers of OT-II cells in flight mice treated with OVA were significantly increased by 2-fold compared with ground mice treated with OVA, suggesting that tolerance induction was impaired by spaceflight. Production of proinflammatory cytokines were significantly increased in flight compared with ground mice, including a 5-fold increase in IFN-γ and a 10-fold increase in IL-17. This study is the first to show that immune tolerance may be impaired in spaceflight, leading to excessive inflammatory responses.
Collapse
Affiliation(s)
- Tammy T Chang
- *Department of Surgery and Department of Medicine, University of California, San Franscisco, San Francisco, California, USA; and Northern California Institute for Research and Education, San Francisco, California, USA
| | - Sandra M Spurlock
- *Department of Surgery and Department of Medicine, University of California, San Franscisco, San Francisco, California, USA; and Northern California Institute for Research and Education, San Francisco, California, USA
| | - Tara Lynne T Candelario
- *Department of Surgery and Department of Medicine, University of California, San Franscisco, San Francisco, California, USA; and Northern California Institute for Research and Education, San Francisco, California, USA
| | - S Marlene Grenon
- *Department of Surgery and Department of Medicine, University of California, San Franscisco, San Francisco, California, USA; and Northern California Institute for Research and Education, San Francisco, California, USA
| | - Millie Hughes-Fulford
- *Department of Surgery and Department of Medicine, University of California, San Franscisco, San Francisco, California, USA; and Northern California Institute for Research and Education, San Francisco, California, USA
| |
Collapse
|
12
|
Abstract
Salmonella are a common source of food- or water-borne infection and cause a wide range of clinical disease in human and animal hosts. Salmonella are relatively easy to culture and manipulate in a laboratory setting, and the infection of laboratory animals induces robust innate and adaptive immune responses. Thus, immunologists have frequently turned to Salmonella infection models to expand understanding of host immunity to intestinal pathogens. In this review, I summarize current knowledge of innate and adaptive immunity to Salmonella and highlight features of this response that have emerged from recent studies. These include the heterogeneity of the antigen-specific T-cell response to intestinal infection, the prominence of microbial mechanisms to impede T- and B-cell responses, and the contribution of non-cognate pathways for elicitation of T-cell effector functions. Together, these different issues challenge an overly simplistic view of host-pathogen interaction during mucosal infection, but also allow deeper insight into the real-world dynamic of protective immunity to intestinal pathogens.
Collapse
Affiliation(s)
- Stephen J McSorley
- Center for Comparative Medicine, Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| |
Collapse
|
13
|
Wang Y, Sui Y, Kato S, Hogg AE, Steel JC, Morris JC, Berzofsky JA. Vaginal type-II mucosa is an inductive site for primary CD8⁺ T-cell mucosal immunity. Nat Commun 2015; 6:6100. [PMID: 25600442 PMCID: PMC4348041 DOI: 10.1038/ncomms7100] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 12/15/2014] [Indexed: 02/07/2023] Open
Abstract
The structured lymphoid tissues are considered the only inductive sites where primary T-cell immune responses occur. The naïve T cells in structured lymphoid tissues, once being primed by antigen-bearing dendritic cells, differentiate into memory T cells and traffic back to the mucosal sites through the bloodstream. Contrary to this belief, here we show that the vaginal type-II mucosa itself, despite the lack of structured lymphoid tissues, can act as an inductive site during primary CD8(+) T-cell immune responses. We provide evidence that the vaginal mucosa supports both the local immune priming of naïve CD8(+) T cells and the local expansion of antigen-specific CD8(+) T cells, thereby demonstrating a different paradigm for primary mucosal T-cell immune induction.
Collapse
Affiliation(s)
- Yichuan Wang
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland 20892, USA
| | - Yongjun Sui
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland 20892, USA
| | - Shingo Kato
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland 20892, USA
| | - Alison E Hogg
- 1] Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland 20892, USA [2] Aeras, 1405 Research Boulevard, Rockville, Maryland 20850, USA
| | - Jason C Steel
- 1] The University of Queensland, Brisbane, Queensland 4120, Australia [2] Gallipoli Medical Research Foundation, Greenslopes, Queensland 4120, Australia
| | - John C Morris
- University of Cincinnati Cancer Institute, Cincinnati, Ohio 45267, USA
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
14
|
|
15
|
Zhang J, De Masi L, John B, Chen W, Schifferli DM. Improved delivery of the OVA-CD4 peptide to T helper cells by polymeric surface display on Salmonella. Microb Cell Fact 2014; 13:80. [PMID: 24898796 PMCID: PMC4055283 DOI: 10.1186/1475-2859-13-80] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 05/19/2014] [Indexed: 12/31/2022] Open
Abstract
Background Autotransporter proteins represent a treasure trove for molecular engineers who modify Gram-negative bacteria for the export or secretion of foreign proteins across two membrane barriers. A particularly promising direction is the development of autotransporters as antigen display or secretion systems. Immunologists have been using ovalbumin as a reporter antigen for years and have developed sophisticated tools to detect specific T cells that respond to ovalbumin. Although ovalbumin-expressing bacteria are being used to trace T cell responses to colonizing or invading pathogens, current constructs for ovalbumin presentation have not been optimized. Results The activation of T helper cells in response to ovalbumin was improved by displaying the OVA-CD4 reporter epitope as a multimer on the surface of Salmonella and fused to the autotransporter MisL. Expression was optimized by including tandem in vivo promoters and two post-segregational killing systems for plasmid stabilization. Conclusions The use of an autotransporter protein to present relevant epitope repeats on the surface of bacteria, combined with additional techniques favoring stable and efficient in vivo transcription, optimizes antigen presentation to T cells. The technique of multimeric epitope surface display should also benefit the development of new Salmonella or other enterobacterial vaccines.
Collapse
Affiliation(s)
| | | | | | | | - Dieter M Schifferli
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania 19104, USA.
| |
Collapse
|
16
|
Ciabattini A, Pettini E, Medaglini D. CD4(+) T Cell Priming as Biomarker to Study Immune Response to Preventive Vaccines. Front Immunol 2013; 4:421. [PMID: 24363656 PMCID: PMC3850413 DOI: 10.3389/fimmu.2013.00421] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 11/20/2013] [Indexed: 01/10/2023] Open
Abstract
T cell priming is a critical event in the initiation of the immune response to vaccination since it deeply influences both the magnitude and the quality of the immune response induced. CD4(+) T cell priming, required for the induction of high-affinity antibodies and immune memory, represents a key target for improving and modulating vaccine immunogenicity. A major challenge in the study of in vivo T cell priming is due to the low frequency of antigen-specific T cells. This review discusses the current knowledge on antigen-specific CD4(+) T cell priming in the context of vaccination, as well as the most advanced tools for the characterization of the in vivo T cell priming and the opportunities offered by the application of systems biology.
Collapse
Affiliation(s)
- Annalisa Ciabattini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Elena Pettini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| | - Donata Medaglini
- Laboratorio di Microbiologia Molecolare e Biotecnologia (LA.M.M.B.), Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, Italy
| |
Collapse
|
17
|
Mittal A, Raber AS, Schaefer UF, Weissmann S, Ebensen T, Schulze K, Guzmán CA, Lehr CM, Hansen S. Non-invasive delivery of nanoparticles to hair follicles: A perspective for transcutaneous immunization. Vaccine 2013; 31:3442-51. [DOI: 10.1016/j.vaccine.2012.12.048] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Revised: 11/23/2012] [Accepted: 12/18/2012] [Indexed: 10/27/2022]
|
18
|
Bacher P, Scheffold A. Flow-cytometric analysis of rare antigen-specific T cells. Cytometry A 2013; 83:692-701. [PMID: 23788442 DOI: 10.1002/cyto.a.22317] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 05/09/2013] [Accepted: 05/14/2013] [Indexed: 12/20/2022]
Abstract
The cytometric enumeration and characterization of antigen-specific lymphocytes, as introduced about 15 years ago, has contributed significantly to our understanding of adaptive immune responses in health and disease. Despite the development of several technologies, allowing to directly or indirectly analyze many aspects of lymphocyte specificity and function, several unresolved issues remain, due to the low frequency of certain antigen-specific lymphocyte subsets and the complexity of T cell antigen recognition. This is especially true for CD4(+) conventional as well as regulatory T cells, which bring major contributions to immune protection and pathology. Here we review the current technologies for the analysis of antigen specific T cells within the physiologic T cell repertoire and with a special focus on recent technologies addressing the analysis of rare antigen-specific T cell populations including naive and regulatory T cells.
Collapse
Affiliation(s)
- Petra Bacher
- Miltenyi Biotec GmbH, Bergisch Gladbach, Germany
| | | |
Collapse
|
19
|
Perforin deficiency impairs a critical immunoregulatory loop involving murine CD8(+) T cells and dendritic cells. Blood 2013; 121:5184-91. [PMID: 23660960 DOI: 10.1182/blood-2013-04-495309] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Humans and mice with impaired perforin-dependent cytotoxic function may develop excessive T-cell activation and the fatal disorder hemophagocytic lymphohistiocytosis (HLH) after infection. Though cytotoxic lymphocytes can kill antigen-presenting cells, the physiological mechanism of perforin-mediated immune regulation has never been demonstrated in a disease-relevant context. We used a murine model of HLH to examine how perforin controls immune activation, and we have defined a feedback loop that is critical for immune homeostasis. This endogenous feedback loop involves perforin-dependent elimination of rare, antigen-presenting dendritic cells (DCs) by CD8(+) T cells and has a dominant influence on the magnitude of T-cell activation after viral infection. Antigen presentation by a minor fraction of DCs persisted in T-cell- or perforin-deficient animals and continued to drive T-cell activation well beyond initial priming in the latter animals. Depletion of DCs or transfer of perforin-sufficient T cells dampened endogenous DC antigen presentation and T-cell activation, demonstrating a reciprocal relationship between perforin in CD8(+) T cells and DC function. Thus, selective cytotoxic "pruning" of DC populations by CD8(+) T cells limits T-cell activation and protects against the development of HLH and potentially other immunopathological conditions.
Collapse
|
20
|
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: 5.1] [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
|
21
|
Sehrawat S, Kirak O, Koenig PA, Isaacson MK, Marques S, Bozkurt G, Simas JP, Jaenisch R, Ploegh HL. CD8(+) T cells from mice transnuclear for a TCR that recognizes a single H-2K(b)-restricted MHV68 epitope derived from gB-ORF8 help control infection. Cell Rep 2012; 1:461-71. [PMID: 22832272 DOI: 10.1016/j.celrep.2012.03.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Revised: 03/07/2012] [Accepted: 03/15/2012] [Indexed: 12/22/2022] Open
Abstract
To study the CD8(+) T cell response against a mouse γ-herpes virus, we generated K(b)-MHV-68-ORF8(604-612)RAG(-/-) CD8(+) T cell receptor transnuclear (TN) mice as a source of virus-specific CD8(+) T cells. K(b)-ORF8-Tet(+) CD8(+) T cells, expanded in the course of a resolving MHV-68 infection, served as a source of nucleus donors. Various in vivo and ex vivo assay criteria demonstrated the fine specificity and functionality of TN cells. TN cells proliferated extensively in response to viral infection, helped control viral burden, and exhibited a phenotype similar to that of endogenous K(b)-ORF8-Tet(+) cells. When compared to OT-1 cells, TN cells displayed distinct properties in response to lymphopenia and cognate antigen stimulation, which may be attributable to the affinity of the TCR expressed by the TN cells. The availability of MHV-68-specific CD8(+) TCR TN mice provides a new tool for investigating aspects of host-pathogen interactions unique to γ-herpes viruses.
Collapse
Affiliation(s)
- Sharvan Sehrawat
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, 9 Cambridge Center, Cambridge, MA 02142, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Abstract
Lineage-committed effector CD4(+) T cells are generated at the peak of the primary response and are followed by heterogeneous populations of central and effector memory cells. Here we review the evidence that T helper type 1 (T(H)1) effector cells survive the contraction phase of the primary response and become effector memory cells. We discuss the applicability of this idea to the T(H)2 cell, T(H)17 helper T cell, follicular helper T cell (T(FH) cell) and induced regulatory T cell lineages. We also discuss how central memory cells are formed, with an emphasis on the role of B cells in this process.
Collapse
|
23
|
McGrath MA, Morton AM, Harnett MM. Laser scanning cytometry: capturing the immune system in situ. Methods Cell Biol 2011; 102:231-60. [PMID: 21704841 DOI: 10.1016/b978-0-12-374912-3.00009-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Until recently, it has not been possible to image and functionally correlate the key molecular and cellular events underpinning immunity and tolerance in the intact immune system. Certainly, the field has been revolutionized by the advent of tetramers to identify physiologically relevant specificities of T cells, and the introduction of models in which transgenic T-cell receptor and/or B-cell receptor-bearing lymphocytes are adoptively transferred into normal mice and can then be identified by clonotype-specific antibodies using flow cytometry in vitro, or immunohistochemistry ex vivo. However, these approaches do not allow for quantitative analysis of the precise anatomical, phenotypic, signaling, and functional parameters required for dissecting the development of immune responses in health and disease in vivo. Traditionally, assessment of signal transduction pathways has required biochemical or molecular biological analysis of isolated and highly purified subsets of immune system cells. Inevitably, this creates potential artifacts and does not allow identification of the key signaling events for individual cells present in their microenvironment in situ. These difficulties have now been overcome by new methodologies in cell signaling analysis that are sufficiently sensitive to detect signaling events occurring in individual cells in situ and the development of technologies such as laser scanning cytometry that provide the tools to analyze physiologically relevant interactions between molecules and cells of the innate and the adaptive immune system within their natural environmental niche in vivo.
Collapse
Affiliation(s)
- Mairi A McGrath
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary & Life Sciences, Glasgow Biomedical Research Centre, University of Glasgow, Scotland, UK
| | | | | |
Collapse
|
24
|
Sun LZ, Elsayed S, Aasen TB, Van Do T, Aardal NP, Florvaag E, Vaali K. Comparison between Ovalbumin and Ovalbumin Peptide 323-339 Responses in Allergic Mice: Humoral and Cellular Aspects. Scand J Immunol 2010; 71:329-35. [DOI: 10.1111/j.1365-3083.2010.02382.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
25
|
Chandran SS, Verhoeven D, Teijaro JR, Fenton MJ, Farber DL. TLR2 engagement on dendritic cells promotes high frequency effector and memory CD4 T cell responses. THE JOURNAL OF IMMUNOLOGY 2010; 183:7832-41. [PMID: 19933854 DOI: 10.4049/jimmunol.0901683] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Ligation of TLR by distinct pathogen components provides essential signals for T cell priming, although how individual TLR engagement affects primary and memory T cell responses is not well defined. In this study, we demonstrate distinct effects of TLR2 vs TLR4 engagement on primary and memory CD4 T cell responses due to differential effects on APC. Priming of influenza hemagglutinin (HA)-specific naive CD4 T cells with HA peptide and the TLR2 agonist Pam3CysK in vivo resulted in a high frequency of activated HA-specific CD4 T cells that predominantly produced IL-2 and IL-17, whereas priming with HA peptide and the TLR4 agonist LPS yielded a lower frequency of HA-specific CD4 T cells and predominant IFN-gamma producers. TLR2 agonist priming depended on TLR2 expression by APC, as wild-type CD4 T cells did not expand in response to peptide and Pam3CysK in TLR2-deficient hosts. TLR2-mediated priming also led to an increased frequency of Ag-specific memory CD4 T cells compared with TLR4 priming and mediated enhanced secondary responses to influenza challenge. Our results show that TLR engagement on APC influences both primary and secondary CD4 T cell responses, and suggest that long-term functional capacities of T cells are set by innate signals during early phases of an infection.
Collapse
Affiliation(s)
- Smita S Chandran
- Department of Surgery and Microbiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | | | |
Collapse
|
26
|
Rodríguez-Fernández JL, Riol-Blanco L, Delgado-Martín C. What is the function of the dendritic cell side of the immunological synapse? Sci Signal 2010; 3:re2. [PMID: 20086241 DOI: 10.1126/scisignal.3105re2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The adaptive immune response requires the formation of a specialized interface called the immunological synapse (IS), which is formed between a mature dendritic cell (DC) and a CD4(+) T cell in the lymph node. The IS involves organized motifs formed by cell-surface and cytoplasmic molecules at both the DC side (IS-DC) and the T cell side (IS-T) of the IS. Most studies of the functions of the IS have focused on the IS-T; however, to understand the function(s) of the entire IS, it is also necessary to gain insight into the role(s) of the IS-DC. Unlike T cells, which upon their activation leave the lymph node and return to the circulation, DCs largely become apoptotic and die in the node region. This latter observation and the known stability of the IS, which may last for hours, is consistent with the hypothesis that one of the functions of the IS-DC could be the temporal inhibition of the apoptosis of DCs, which would enable the activation of clonal T cells in the lymph nodes. Here, we discuss experimental data supporting the latter hypothesis, as well as the concept that the IS-DC is a signaling region that contributes to the functions of the IS.
Collapse
Affiliation(s)
- José Luis Rodríguez-Fernández
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, C/ Ramiro de Maeztu, 9, 28040 Madrid, Spain.
| | | | | |
Collapse
|
27
|
Boppana VD, Thangamani S, Alarcon-Chaidez FJ, Adler AJ, Wikel SK. Blood feeding by the Rocky Mountain spotted fever vector, Dermacentor andersoni, induces interleukin-4 expression by cognate antigen responding CD4+ T cells. Parasit Vectors 2009; 2:47. [PMID: 19814808 PMCID: PMC2764639 DOI: 10.1186/1756-3305-2-47] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2009] [Accepted: 10/08/2009] [Indexed: 11/10/2022] Open
Abstract
Background Tick modulation of host defenses facilitates both blood feeding and pathogen transmission. Several tick species deviate host T cell responses toward a Th2 cytokine profile. The majority of studies of modulation of T cell cytokine expression by ticks were performed with lymphocytes from infested mice stimulated in vitro with polyclonal T cell activators. Those reports did not examine tick modulation of antigen specific responses. We report use of a transgenic T cell receptor (TCR) adoptive transfer model reactive with influenza hemagglutinin peptide (110-120) to examine CD4+ T cell intracellular cytokine responses during infestation with the metastriate tick, Dermacentor andersoni, or exposure to salivary gland extracts. Results Infestation with pathogen-free D. andersoni nymphs or administration of an intradermal injection of female or male tick salivary gland extract induced significant increases of IL-4 transcripts in skin and draining lymph nodes of BALB/c mice as measured by quantitative real-time RT-PCR. Furthermore, IL-10 transcripts were significantly increased in skin while IL-2 and IFN-γ transcripts were not significantly changed by tick feeding or intradermal injection of salivary gland proteins, suggesting a superimposed Th2 response. Infestation induced TCR transgenic CD4+ T cells to divide more frequently as measured by CFSE dilution, but more notably these CD4+ T cells also gained the capacity to express IL-4. Intracellular levels of IL-4 were significantly increased. A second infestation administered 14 days after a primary exposure to ticks resulted in partially reduced CFSE dilution with no change in IL-4 expression when compared to one exposure to ticks. Intradermal inoculation of salivary gland extracts from both male and female ticks also induced IL-4 expression. Conclusion This is the first report of the influence of a metastriate tick on the cytokine profile of antigen specific CD4+ T cells. Blood feeding by D. andersoni pathogen-free nymphs or intradermal injection of salivary gland extracts programs influenza hemagglutinin influenza peptide specific TCR transgenic CD4+ T cells to express IL-4.
Collapse
Affiliation(s)
- Venkata D Boppana
- Department of Pathology, Center for Biodefense and Emerging Infectious Diseases and Center for Tropical Diseases, School of Medicine, University of Texas Medical Branch, Galveston, Texas, 77555, USA.
| | | | | | | | | |
Collapse
|
28
|
Abstract
T cells must integrate multiple environmental cues when deciding whether to mount an immunogenic or tolerogenic response. Since not all self-reactive T cells are eliminated during thymic development, mechanisms of peripheral tolerance such as T cell anergy contribute to preventing autoimmunity. Recent studies have implicated extracellular adenosine and the adenosine A(2A) receptor as playing an important role in inhibiting T cell effector function. Herein, we review the current literature regarding T cell anergy and the emerging literature implicating the A(2A) receptor as critical regulator of immune activation. Finally, we present evidence to suggest a possible role for adenosine A(2A) receptor signaling in T cell anergy.
Collapse
Affiliation(s)
- Paul E Zarek
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21231, USA
| | | |
Collapse
|
29
|
Moon JJ, Chu HH, Hataye J, Pagán AJ, Pepper M, McLachlan JB, Zell T, Jenkins MK. Tracking epitope-specific T cells. Nat Protoc 2009; 4:565-81. [PMID: 19373228 DOI: 10.1038/nprot.2009.9] [Citation(s) in RCA: 226] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The tracking of antigen-specific T cells in vivo is a useful approach for the study of the adaptive immune response. This protocol describes how populations of T cells specific for a given peptide-major histocompatibility complex (pMHC) epitope can be tracked based solely on T-cell receptor (TCR) specificity as opposed to other indirect methods based on function. The methodology involves the adoptive transfer of TCR transgenic T cells with defined epitope specificity into histocompatible mice and the subsequent detection of these cells through the use of congenic or clonotypic markers. Alternatively, endogenous epitope-specific T cells can be tracked directly through the use of pMHC tetramers. Using magnetic bead-based enrichment and advanced multiparameter flow cytometry, populations as small as five epitope-specific T cells can be detected from the peripheral lymphoid organs of a mouse. The adoptive transfer procedure can be completed within 3 h, whereas analysis of epitope-specific cells from mice can be completed within 6 h.
Collapse
Affiliation(s)
- James J Moon
- Department of Microbiology and Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Vendetti S, Riccomi A, Negri DRM, Veglia F, Sciaraffia E, De Magistris MT. Development of antigen-specific T cells in mediastinal lymph nodes after intranasal immunization. Methods 2009; 49:334-9. [PMID: 19426804 DOI: 10.1016/j.ymeth.2009.04.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2009] [Revised: 04/16/2009] [Accepted: 04/23/2009] [Indexed: 10/20/2022] Open
Abstract
The power of cholera toxin (CT) as an effective mucosal adjuvant is well established. Because of the high toxicity of CT, its clinical use is unlikely. Therefore, the need to identify effective and non toxic mucosal adjuvants for human use is important. For this purpose, CT is largely used as a reference molecule for testing the efficacy of new candidate adjuvants in animal models. Here, we evaluated the kinetics and the localization of antigen-specific humoral and cellular immune responses elicited by intranasal immunization with tetanus toxoid antigen in the presence of CT. We show that an antigen-specific cellular immune response localized in the mediastinal lymph nodes can be observed already 1 week after the first immunization. The induction of an appreciable titer of an antibody-specific immune response was assessed after two immunizations. Therefore, we suggest that the efficacy of new candidate mucosal adjuvants can be tested by evaluating the cellular immune response in the mediastinal lymph nodes at early stages of immunization.
Collapse
Affiliation(s)
- Silvia Vendetti
- Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | | | | | | | | | | |
Collapse
|
31
|
Pettini E, Ciabattini A, Pozzi G, Medaglini D. Adoptive transfer of transgenic T cells to study mucosal adjuvants. Methods 2009; 49:340-5. [PMID: 19409994 DOI: 10.1016/j.ymeth.2009.03.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2009] [Revised: 02/24/2009] [Accepted: 03/09/2009] [Indexed: 10/20/2022] Open
Abstract
The study of the initiation and regulation of T-cell responses to vaccine antigens is of primary importance in the rational design of mucosal adjuvants. The detection in vivo of T-cell priming following immunization can be performed by using the adoptive transfer model of naïve antigen-specific transgenic T cells into immunocompetent mice. In this work, we discuss the applications of this system for detecting in vivo the primary antigen-specific clonal expansion, the phenotype, and the effector function of transgenic T cells following mucosal immunization. OVA and the mucosal adjuvant CTB were used as a model vaccine formulation and administered by the nasal route to study T-cell priming. T helper and T cytotoxic primary proliferation and expression of activation and migration markers was observed both in draining and distal sites. This method proved to be a powerful tool to study the efficacy of mucosal adjuvants in enhancing T-cell priming.
Collapse
Affiliation(s)
- Elena Pettini
- Laboratorio di Microbiologia Molecolare e Biotecnologia, Dipartimento di Biologia Molecolare, Università di Siena, 53100 Siena, Italy
| | | | | | | |
Collapse
|
32
|
Abstract
Over the last decade, significant advances have been made in the methodology for studying immune responses in vivo. It is now possible to follow almost every aspect of pathogen-specific immunity using in vivo models that incorporate physiological infectious doses and natural routes of infection. This new ability to study immunity in a relevant physiological context will greatly expand our understanding of the dynamic interplay between host and pathogen. Visualizing the resolution of primary infection and the development of long-term immunological memory should also aid the development of new vaccines and therapeutics for infectious diseases. In this review, we will describe the application of in vivo visualization technology to Salmonella infection, describe our current understanding of Salmonella-specific immunity, and discuss some unanswered questions that remain in this model.
Collapse
Affiliation(s)
- James J. Moon
- Department of Microbiology, University of Minnesota Medical School, Minneapolis, MN, 55455
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455
| | - Stephen J. McSorley
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, 55455
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455
- Center for Infectious Diseases & Microbiology Translational Research, University of Minnesota Medical School, Minneapolis, MN, 55455
| |
Collapse
|
33
|
Duffy D, Sparshott SM, Yang CP, Bell EB. Transgenic CD4 T cells (DO11.10) are destroyed in MHC-compatible hosts by NK cells and CD8 T cells. THE JOURNAL OF IMMUNOLOGY 2008; 180:747-53. [PMID: 18178812 DOI: 10.4049/jimmunol.180.2.747] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
During an immune response a small number of rare Ag-specific clones proliferate extensively and decline, leaving a residual population for long-term memory. TCR transgenic (tg) CD4 T cells have been used widely to study the primary and memory response in vivo. We show here that naive TCR tg CD4 T cells from the DO11.10 strain transferred into wild type (wt) BALB/c recipients and not stimulated declined rapidly at the same rate as those primed in vivo by Ag. In the same recipients wt CD4 T cells survived. There was no evidence of an inherent defect in the tg T cells, which survived well when returned to DO11.10 recipients. Surprisingly, wt CD4 T cells declined rapidly in the same DO11.10 hosts. By depleting wt recipients of NK cells or CD8+ cells, the speed of reduction was slowed by half; rapid destruction was prevented completely by combing the two treatments. In contrast, preimmunization accelerated the loss of tg T cells. The results suggested that tg CD4 T cells were actively rejected by both NK and CD8 T cell responses. We consider whether, despite extensive backcrossing, tg T cells may retain genetic material (minor histocompatibility Ags) flanking the construct that compromises their survival in wt recipients.
Collapse
Affiliation(s)
- Darragh Duffy
- Immunology Section, Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | | | | | | |
Collapse
|
34
|
Yi KH, Nechushtan H, Bowers WJ, Walker GR, Zhang Y, Pham DG, Podack ER, Federoff HJ, Tolba KA, Rosenblatt JD. Adoptively transferred tumor-specific T cells stimulated ex vivo using herpes simplex virus amplicons encoding 4-1BBL persist in the host and show antitumor activity in vivo. Cancer Res 2007; 67:10027-37. [PMID: 17942937 DOI: 10.1158/0008-5472.can-06-2391] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
4-1BB is a T-cell costimulatory receptor which binds its ligand 4-1BBL, resulting in prolonged T cell survival. We studied the antitumor effects of adoptively transferred tumor-specific T cells expanded ex vivo using tumors transduced with herpes simplex virus (HSV) amplicons expressing 4-1BBL as a direct source of antigen and costimulation. We constructed HSV amplicons encoding either the 4-1BBL (HSV.4-1BBL) or B7.1 (HSV.B7.1) costimulatory ligands. Lewis lung carcinoma cells expressing ovalbumin (LLC/OVA) were transduced with HSV.4-1BBL, HSV.B7.1, or control HSV amplicons and used to stimulate GFP+ OVA-specific CD8+ T cells (OT-1/GFP) ex vivo. Naive or ex vivo stimulated OT-1/GFP cells were adoptively transferred into LLC/OVA tumor-bearing mice. Higher percentages of OT-1/GFP cells were seen in the peripheral blood, spleen, and tumor bed of the HSV.4-1BBL-stimulated OT-1/GFP group compared with all other experimental groups. OT-1 cells identified within the tumor bed and draining lymph nodes of the HSV.4-1BBL-stimulated OT-1 group showed enhanced bromodeoxyuridine (BrdUrd) incorporation, suggesting ongoing expansion in vivo. Mice receiving HSV.4-1BBL-stimulated OT-1/GFP had significantly decreased tumor volumes compared with untreated mice (P<0.001) or to mice receiving naive OT-1/GFP (P<0.001). Transfer of HSV.B7.1-stimulated OT-1/GFP did not protect mice from tumor. Mice that received HSV.4-1BBL-stimulated OT-1/GFP exhibited increased cytolytic activity against LLC/OVA and higher percentages of Ly-6C+ OT-1/GFP in the spleen and tumor bed compared with controls. Tumor-specific T cells stimulated ex vivo using tumor transduced with HSV.4-1BBL expand in vivo following adoptive transfer, resulting in tumor eradication and the generation of tumor-specific CD44+Ly-6C+CD62L- effector memory T cells.
Collapse
Affiliation(s)
- Kyung H Yi
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, and University of Miami Sylvester Comprehensive Cancer Center, Miami, Florida 33136, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
|
36
|
Iancu EM, Speiser DE, Rufer N. Assessing ageing of individual T lymphocytes: mission impossible? Mech Ageing Dev 2007; 129:67-78. [PMID: 18048082 DOI: 10.1016/j.mad.2007.10.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2007] [Revised: 10/15/2007] [Accepted: 10/23/2007] [Indexed: 12/22/2022]
Abstract
Effector T lymphocytes are the progeny of a limited number of antigen-specific precursor cells and it has been estimated that clonotypic human T cells may expand million fold on their way reaching high cell numbers that are sufficient for immune protection. Moreover, memory T cell responses are characterized by repetitive expansion of antigen-specific T cell clonotypes, and limitations in the proliferative capacity could lead to immune senescence. Because telomeres progressively shorten as a function of cell division, telomere length is a powerful indicator of the replicative in vivo history of human T lymphocytes. In this review, we summarize observations made over the last decade on telomere length dynamics of well-defined T cell populations derived from healthy donors and patients with infectious disease or cancer. We focus on T cell differentiation, T cell ageing, and natural and vaccine induced immune responses. We also discuss the scientific evidence for in vivo replicative senescence of antigen-specific T cells, and evaluate the available methods for measuring telomere lengths and telomerase activity, and their potential and limitations to increase our understanding of T cell physiology.
Collapse
Affiliation(s)
- Emanuela M Iancu
- Division of Experimental Oncology, Multidisciplinary Oncology Center CePO, Avenue Pierre-Decker 4, CH-1005 Lausanne, Switzerland
| | | | | |
Collapse
|
37
|
Marshall FA, Watson KA, Garside P, Harnett MM, Harnett W. Effect of activated antigen-specific B cells on ES-62-mediated modulation of effector function of heterologous antigen-specific T cells in vivo. Immunology 2007; 123:411-25. [PMID: 17961164 PMCID: PMC2433340 DOI: 10.1111/j.1365-2567.2007.02706.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
There is currently great interest in the idea of using helminth-derived molecules for therapeutic purposes and indeed we have shown that ES-62, a filarial nematode-derived phosphorylcholine-containing glycoprotein, significantly reduces the severity of arthritis in a murine model. Clearly, knowledge of mechanism of action is important when considering molecules for use in treating disease and although much is known regarding how ES-62 interacts with the immune system, gaps in our understanding remain. A feature of filarial nematode infection is a defective, T helper 2 (Th2)-polarized antigen-specific T-cell response and in relation to this we have recently shown that ES-62 inhibits clonal expansion and modulates effector function towards a Th2 phenotype, of antigen-specific T cells in vivo. ES-62 is also known to directly modulate B-cell behaviour and hence to determine whether it was mediating these effects on T cells by disrupting B-T-cell co-operation, we have investigated antigen-specific responses using an adoptive transfer system in which traceable numbers of tg ovalbumin (OVA)-specific T cells and hen egg lysozyme (HEL)-specific B cells respond to a chemically coupled form of OVA-HEL that contains linked epitopes that promote cognate T- and B-cell interactions. Surprisingly, these studies indicate that activated B cells restore T-cell expansion and prevent Th2-like polarization. However, ES-62-treated double cell transfer mice demonstrate a more generalized immunosuppression with reduced levels of Th1 and -2 type cytokines and antibody subclasses. Collectively, these results suggest that whilst ES-62 can target B-T-cell co-operation, this does not promote polarizing of T-cell responses towards a Th2-type phenotype.
Collapse
Affiliation(s)
- Fraser A Marshall
- Division of Immunology, Infection and Inflammation, University of Glasgow, Glasgow, UK
| | | | | | | | | |
Collapse
|
38
|
Müller-Doblies UU, Maxwell SS, Boppana VD, Mihalyo MA, McSorley SJ, Vella AT, Adler AJ, Wikel SK. Feeding by the tick, Ixodes scapularis, causes CD4+T cells responding to cognate antigen to develop the capacity to express IL-4. Parasite Immunol 2007; 29:485-99. [PMID: 17883452 DOI: 10.1111/j.1365-3024.2007.00966.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Effects of tick feeding on an early antigen-specific T cell response were studied by monitoring a clonotypic population of adoptively transferred T cell receptor (TCR) transgenic CD4 cells responding to a tick-associated antigen. When recipient mice were infested with pathogen-free Ixodes scapularis nymphs several days prior to T cell transfer and intradermal injection of soluble cognate antigen at the feeding site, the clonotypic CD4 cells gained the ability to express the Th2 effector cytokine IL-4. Notably, this effect was not only observed in BALB/c mice predisposed towards developing Th2 responses but also in B10.D2 mice predisposed towards Th1 responsiveness. Furthermore, tick feeding was able to superimpose IL-4 expression potential onto a strong Th1 response (indicated by robust IFN-gamma expression potential) elicited by immunization with a vaccinia virus expressing the cognate antigen. The magnitude to which tick feeding was able to programme IL-4 expression potential in CD4 cells was partially reduced in mice that had been previously exposed to pathogen-free tick nymphs 6 weeks earlier, as well as when the nymphs were infected with Borrelia burgdorferi. Intradermal injection of salivary gland extract programmed IL-4 expression potential similar to that of tick infestation, suggesting that IL-4 programming activity is contained within tick saliva.
Collapse
|
39
|
Kirchhoff D, Frentsch M, Leclerk P, Bumann D, Rausch S, Hartmann S, Thiel A, Scheffold A. Identification and isolation of murine antigen-reactive T cells according to CD154 expression. Eur J Immunol 2007; 37:2370-7. [PMID: 17705136 DOI: 10.1002/eji.200737322] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
T helper (Th) cells are central regulators of adaptive immune responses. However, the detection of the small number of Th cells specific for a particular antigen or pathogen is still a major challenge. CD154 was recently introduced as a marker for antigen-specific Th cells. To date, this technology was not applicable for mice - arguably the most important immunological model system. CD154 is difficult to detect due to its rapid removal from the cell surface upon binding to CD40 during antigen-specific activation by APC. We present an efficient strategy to block the degradation of murine CD154 by combined use of antibodies against CD40 and CD154. This strategy makes CD154 easily accessible for surface staining, which allows isolation and expansion of rare antigen specific T cells. Importantly, CD154 identified all specific T cells in strongly Th1- or Th2-polarized immune responses against pathogens like Salmonella typhimurium and Heligmosomoides polygyrus, independent of their potential to produce cytokines. We demonstrate that CD154 can in fact be used as a reliable marker for antigen-specific CD4 T cells in mice, offering a unique option to analyze, isolate and rapidly expand the entire pool of Th-cells generated during a physiological T cell response in vivo.
Collapse
Affiliation(s)
- Dennis Kirchhoff
- Immunomodulation Group, Deutsches Rheuma-Forschungszentrum Berlin, Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Mescher MF, Popescu FE, Gerner M, Hammerbeck CD, Curtsinger JM. Activation-induced non-responsiveness (anergy) limits CD8 T cell responses to tumors. Semin Cancer Biol 2007; 17:299-308. [PMID: 17656106 PMCID: PMC2693139 DOI: 10.1016/j.semcancer.2007.06.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2007] [Accepted: 06/07/2007] [Indexed: 12/22/2022]
Abstract
Naïve CD8 T cells respond to signals provided by Ag, costimulation and cytokines by proliferating and differentiating to develop effector functions. Following initial clonal expansion, however, the cells develop activation-induced non-responsiveness (AINR), a form of anergy characterized by an inability to produce IL-2. Cells in the AINR state can carry out effector functions (cytolysis, IFN-gamma production) but cannot continue to proliferate and expand in the face of persisting Ag. AINR limits the ability of activated CTL to control tumor growth but can be reversed by IL-2, provided either therapeutically or by activated CD4 T helper cells, to allow continued expansion.
Collapse
Affiliation(s)
- Matthew F Mescher
- Center for Immunology and Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware Street S.E., Minneapolis, MN 55455, USA.
| | | | | | | | | |
Collapse
|
41
|
Costalonga M, Zell T. Lipopolysaccharide enhances in vivo interleukin-2 production and proliferation by naive antigen-specific CD4 T cells via a Toll-like receptor 4-dependent mechanism. Immunology 2007; 122:124-30. [PMID: 17484770 PMCID: PMC2265990 DOI: 10.1111/j.1365-2567.2007.02620.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Microbial adjuvants are essential for the development of T-cell-dependent antibody production, recall T-cell proliferation and interferon-gamma production following immunization with protein antigens. Using an adoptive transfer approach, we showed that the adjuvant lipopolysaccharide enhanced the frequency of cells producing interleukin-2, enhanced clonal expansion by antigen-specific CD4 T cells and increased CD86 and interleukin-1alpha production by antigen-presenting cells. All of these effects were dependent on Toll-like receptor-4 (TLR4) expression by cells other than the antigen-specific CD4 T cells. The ability of lipopolysaccharides to increase the number of antigen-specific CD4 T cells that survive after immunization probably explains the previous finding that antigen-specific proliferation by T cells from normal mice depends on previous exposure to antigen and adjuvant.
Collapse
Affiliation(s)
- Massimo Costalonga
- Department of Developmental and Surgical Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA.
| | | |
Collapse
|
42
|
Zheng Y, Collins SL, Lutz MA, Allen AN, Kole TP, Zarek PE, Powell JD. A role for mammalian target of rapamycin in regulating T cell activation versus anergy. THE JOURNAL OF IMMUNOLOGY 2007; 178:2163-70. [PMID: 17277121 DOI: 10.4049/jimmunol.178.4.2163] [Citation(s) in RCA: 209] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Whether TCR engagement leads to activation or tolerance is determined by the concomitant delivery of multiple accessory signals, cytokines, and environmental cues. In this study, we demonstrate that the mammalian target of rapamycin (mTOR) integrates these signals and determines the outcome of TCR engagement with regard to activation or anergy. In vitro, Ag recognition in the setting of mTOR activation leads to full immune responses, whereas recognition in the setting of mTOR inhibition results in anergy. Full T cell activation is associated with an increase in the phosphorylation of the downstream mTOR target S6 kinase 1 at Thr(421)/Ser(424) and an increase in the mTOR-dependent cell surface expression of transferrin receptor (CD71). Alternatively, the induction of anergy results in markedly less S6 kinase 1 Thr(421)/Ser(424) phosphorylation and CD71 surface expression. Likewise, the reversal of anergy is associated not with proliferation, but rather the specific activation of mTOR. Importantly, T cells engineered to express a rapamycin-resistant mTOR construct are resistant to anergy induction caused by rapamycin. In vivo, mTOR inhibition promotes T cell anergy under conditions that would normally induce priming. Furthermore, by examining CD71 surface expression, we are able to distinguish and differentially isolate anergic and activated T cells in vivo. Overall, our data suggest that by integrating environmental cues, mTOR plays a central role in determining the outcome of Ag recognition.
Collapse
Affiliation(s)
- Yan Zheng
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University Medical Institutions, Baltimore, MD 21231, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Duffy D, Yang CP, Heath A, Garside P, Bell EB. Naive T-cell receptor transgenic T cells help memory B cells produce antibody. Immunology 2007; 119:376-84. [PMID: 17067314 PMCID: PMC1819572 DOI: 10.1111/j.1365-2567.2006.02446.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Injection of the same antigen following primary immunization induces a classic secondary response characterized by a large quantity of high-affinity antibody of an immunoglobulin G class produced more rapidly than in the initial response - the products of memory B cells are qualitatively distinct from that of the original naive B lymphocytes. Very little is known of the help provided by the CD4 T cells that stimulate memory B cells. Using antigen-specific T-cell receptor transgenic CD4 T cells (DO11.10) as a source of help, we found that naive transgenic T cells stimulated memory B cells almost as well (in terms of quantity and speed) as transgenic T cells that had been recently primed. There was a direct correlation between serum antibody levels and the number of naive transgenic T cells transferred. Using T cells from transgenic interleukin-2-deficient mice we showed that interleukin-2 was not required for a secondary response, although it was necessary for a primary response. The results suggested that the signals delivered by CD4 T cells and required by memory B cells for their activation were common to both antigen-primed and naive CD4 T cells.
Collapse
Affiliation(s)
- Darragh Duffy
- Division of Immunology, Faculty of Life Sciences, The University of Manchester, Manchester, UK.
| | | | | | | | | |
Collapse
|
44
|
Roan NR, Starnbach MN. Antigen-specific CD8+ T cells respond to Chlamydia trachomatis in the genital mucosa. THE JOURNAL OF IMMUNOLOGY 2007; 177:7974-9. [PMID: 17114470 DOI: 10.4049/jimmunol.177.11.7974] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Following sexual transmission, Chlamydia trachomatis specifically targets genital tract epithelial cells. Because epithelial cells are readily recognized by CD8+ T cells, the response of CD8+ T cells to Chlamydia infection has been explored in a number of studies. It has been shown that CD8+ T cells are present in the genital tracts of mice following C. trachomatis infection, but the specificity of these T cells has remained undefined. To determine whether Chlamydia-specific CD8+ T cells migrate to the genital tract in response to Chlamydia infection, we generated retrogenic mice that express a TCR specific for a Chlamydia-specific T cell Ag CrpA. T cells from the retrogenic mice were transferred into naive recipient animals to increase the frequency of Chlamydia-specific T cells to a level at which they could be tracked during primary infection. We observed that the Chlamydia-specific retrogenic T cells proliferated in lymph nodes draining the genital tract in response to genital infection with C. trachomatis. Furthermore, we found that these cells acquired the ability to produce IFN-gamma and migrated into the genital mucosa of the infected mice.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigens, Bacterial/genetics
- Antigens, Bacterial/immunology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/microbiology
- Chemotaxis, Leukocyte/immunology
- Chlamydia Infections/immunology
- Chlamydia trachomatis/immunology
- Female
- Flow Cytometry
- Gene Transfer Techniques
- Genitalia, Female/immunology
- Genitalia, Female/microbiology
- Interferon-gamma/immunology
- Interferon-gamma/metabolism
- Lymph Nodes/cytology
- Lymph Nodes/immunology
- Mice
- Mice, Inbred C57BL
- Mucous Membrane/cytology
- Mucous Membrane/immunology
- Mucous Membrane/microbiology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
Collapse
Affiliation(s)
- Nadia R Roan
- Department of Microbiology and Molecular Genetics, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA
| | | |
Collapse
|
45
|
Tse HM, Milton MJ, Schreiner S, Profozich JL, Trucco M, Piganelli JD. Disruption of Innate-Mediated Proinflammatory Cytokine and Reactive Oxygen Species Third Signal Leads to Antigen-Specific Hyporesponsiveness. THE JOURNAL OF IMMUNOLOGY 2007; 178:908-17. [PMID: 17202352 DOI: 10.4049/jimmunol.178.2.908] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Successful Ag activation of naive T helper cells requires at least two signals consisting of TCR and CD28 on the T cell interacting with MHC II and CD80/CD86, respectively, on APCs. Recent evidence demonstrates that a third signal consisting of proinflammatory cytokines and reactive oxygen species (ROS) produced by the innate immune response is important in arming the adaptive immune response. In an effort to curtail the generation of an Ag-specific T cell response, we targeted the synthesis of innate immune response signals to generate Ag-specific hyporesponsiveness. We have reported that modulation of redox balance with a catalytic antioxidant effectively inhibited the generation of third signal components from the innate immune response (TNF-alpha, IL-1beta, ROS). In this study, we demonstrate that innate immune-derived signals are necessary for adaptive immune effector function and disruption of these signals with in vivo CA treatment conferred Ag-specific hyporesponsiveness in BALB/c, NOD, DO11.10, and BDC-2.5 mice after immunization. Modulating redox balance led to decreased Ag-specific T cell proliferation and IFN-gamma synthesis by diminishing ROS production in the APC, which affected TNF-alpha levels produced by CD4(+) T cells and impairing effector function. These results demonstrate that altering redox status can be effective in T cell-mediated diseases such as autoimmune diabetes to generate Ag-specific immunosuppression because it inhibits the third signal necessary for CD4(+) T cells to transition from expansion to effector function.
Collapse
Affiliation(s)
- Hubert M Tse
- Diabetes Institute, Division of Immunogenetics, Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | | | | | | | |
Collapse
|
46
|
Hamdy S, Elamanchili P, Alshamsan A, Molavi O, Satou T, Samuel J. Enhanced antigen-specific primary CD4+ and CD8+ responses by codelivery of ovalbumin and toll-like receptor ligand monophosphoryl lipid A in poly(D,L-lactic-co-glycolic acid) nanoparticles. J Biomed Mater Res A 2007; 81:652-62. [PMID: 17187395 DOI: 10.1002/jbm.a.31019] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The purpose of this research was to investigate the use of biodegradable poly(D,L-lactic-co-glycolic acid) nanoparticles (PLGA-NP) as a vaccine delivery system to codeliver antigen, ovalbumin (OVA) along with monophosphoryl lipid A (MPLA) as adjuvant for induction of potent CD4(+) and CD8(+) T cell responses. The primary CD4(+) T responses to OVA/MPLA NP were investigated using OVA-specific T cells from DO11.10 transgenic mice. Following adoptive transfer of these cells, mice were immunized s.c. by NP formulations. For assessing the CD8(+) responses, bone marrow derived dendritic cells (DCs) were pulsed with different OVA formulations, then, cocultured with CD8(+) T cells from OT-1 mice. T cell proliferation/activation and IFN-gamma secretion profile have been examined. Particulate delivery of OVA and MPLA to the DCs lead to markedly increase in in vitro CD8(+) T cell T cell proliferative responses (stimulation index >3000) and >13-folds increase in in vivo clonal expanded CD4(+) T cells. The expanded T cells were capable of cytokine secretion and expressed an activation and memory surface phenotype (CD62L(lo), CD11a(hi), and CD44(hi)). Codelivery of antigen and MPLA in PLGA-NP offers an effective method for induction of potent antigen specific CD4(+) and CD8(+) T cell responses.
Collapse
Affiliation(s)
- Samar Hamdy
- Faculty of Pharmacy and Pharmaceutical Sciences, 3133 Dentistry/Pharmacy Centre, University of Alberta, Edmonton, Alberta, Canada, T6G 2N8
| | | | | | | | | | | |
Collapse
|
47
|
Verwaerde C, Delanoye A, Macia L, Tailleux A, Wolowczuk I. Influence of high-fat feeding on both naive and antigen-experienced T-cell immune response in DO10.11 mice. Scand J Immunol 2006; 64:457-66. [PMID: 17032237 DOI: 10.1111/j.1365-3083.2006.01791.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Obesity is becoming one of the most serious public health problems in industrialized societies, due to the profound changes in lifestyle, and notably in nutrition. Beside diabetes, cardiovascular diseases or hypertension, increased susceptibility to infection is one of the pathological consequences of being overweight. In this paper, we have assessed the influence of a high-fat diet (HFD) rich in saturated fatty acids on the immune system of DO11.10 mice, which are transgenic for a T-cell receptor specifically recognizing a peptide of ovalbumin. We showed that the specific T-cell immune response was impaired by high-fat feeding, and that the expression of this defect is different depending on whether T cells are naive or Ag experienced. Indeed, on in vitro ovalbumin stimulation, spleen T cells from naive HFD-fed transgenic mice showed proliferation similar to that of cells from standard diet (SD)-fed mice, but exhibited a strong inflammatory profile as shown by the markedly increased IFN-gamma/IL-4 ratio. Inversely, spleen T cells from ovalbumin-immunized HFD mice were impaired in their Ag-dependent proliferation compared to cells from SD mice. By co-culture experiments, we showed that both T cells and antigen-presenting cells were involved in this impairment. Moreover, in ovalbumin-immunized HFD animals, a trend towards Th2 response was noted, compared to immunized SD mice. This data implies that naive T cells could participate actively in the low-grade systemic inflammation observed in overweight patients. Moreover, the impaired activity of Ag-experienced T cells could have major consequences both in defence against infection and/or in vaccination protocols.
Collapse
Affiliation(s)
- C Verwaerde
- UMR-CNRS 8527, Institut de Biologie de Lille/Institut Pasteur de Lille, Université de Lille II, Lille, France.
| | | | | | | | | |
Collapse
|
48
|
Jabbari A, Legge KL, Harty JT. T cell conditioning explains early disappearance of the memory CD8 T cell response to infection. THE JOURNAL OF IMMUNOLOGY 2006; 177:3012-8. [PMID: 16920937 DOI: 10.4049/jimmunol.177.5.3012] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Memory CD8 T cells respond more rapidly to acute intracellular infections than naive CD8 T cells. An understanding of the biological processes involved in memory CD8 T cell recognition of Ag and up-regulation of effector mechanism necessitates analyzing memory CD8 T cells at early time points after infection. In the current study, we show that memory CD8 T cells ostensibly disappear from the spleens, blood, and peripheral organs of mice early after infection with Listeria monocytogenes. This disappearance is critically dependent on Ag, and cell-associated Ag alone can mediate this phenomenon. Further investigations, however, suggest that this disappearance is secondary to T cell-APC interactions, also known as T cell conditioning, and disruption of these putative interactions during splenic processing improves recovery of Ag-specific memory CD8 T cell populations after immunization. Conventional analyses of memory CD8 T cell populations early after infection and possibly in the presence of low levels of Ag (as during chronic infections) may exclude significant numbers of the responding CD8 T cell population.
Collapse
Affiliation(s)
- Ali Jabbari
- Interdisciplinary Graduate Program in Immunology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | | | | |
Collapse
|
49
|
Loeffler DIM, Schoen CU, Goebel W, Pilgrim S. Comparison of different live vaccine strategies in vivo for delivery of protein antigen or antigen-encoding DNA and mRNA by virulence-attenuated Listeria monocytogenes. Infect Immun 2006; 74:3946-57. [PMID: 16790768 PMCID: PMC1489688 DOI: 10.1128/iai.00112-06] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Listeria monocytogenes can be used to deliver protein antigens or DNA and mRNA encoding such antigens directly into the cytosol of host cells because of its intracellular lifestyle. In this study, we compare the in vivo efficiencies of activation of antigen-specific CD8 and CD4 T cells when the antigen is secreted by L. monocytogenes or when antigen-encoding plasmid DNA or mRNA is released by self-destructing strains of L. monocytogenes. Infection of mice with self-destructing L. monocytogenes carriers delivering mRNA that encodes a nonsecreted form of ovalbumin (OVA) resulted in a significant OVA-specific CD8 T-cell response. In contrast, infection with L. monocytogenes delivering OVA-encoding DNA failed to generate specific T cells. Secretion of OVA by the carrier bacteria yielded the strongest immune response involving OVA-specific CD8 and CD4 T cells. In addition, we investigated the antigen delivery capacity of a self-destructing, virulence-attenuated L. monocytogenes aroA/B mutant. In contrast to the wild-type strain, this mutant exhibited only marginal liver toxicity when high doses (5 x 10(7) CFU per animal administered intravenously) were used, and it was also able to deliver sufficient amounts of secreted OVA into mice. Therefore, the results presented here could lay the groundwork for a rational combination of L. monocytogenes as an attenuated carrier for the delivery of protein and nucleic acid vaccines in novel vaccination strategies.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigens, Bacterial/administration & dosage
- Antigens, Bacterial/genetics
- Antigens, Bacterial/immunology
- Bacterial Vaccines/administration & dosage
- Bacterial Vaccines/genetics
- Bacterial Vaccines/immunology
- Cell Line, Tumor
- DNA, Bacterial/administration & dosage
- DNA, Bacterial/immunology
- Drug Delivery Systems
- Female
- Leukemia P388
- Listeria monocytogenes/genetics
- Listeria monocytogenes/immunology
- Listeria monocytogenes/pathogenicity
- Listeriosis/immunology
- Listeriosis/prevention & control
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Ovalbumin/administration & dosage
- Ovalbumin/immunology
- Ovalbumin/metabolism
- RNA, Bacterial/administration & dosage
- RNA, Bacterial/immunology
- RNA, Messenger/administration & dosage
- RNA, Messenger/immunology
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/genetics
- Vaccines, Attenuated/immunology
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/immunology
- Virulence
Collapse
|
50
|
Roan NR, Gierahn TM, Higgins DE, Starnbach MN. Monitoring the T cell response to genital tract infection. Proc Natl Acad Sci U S A 2006; 103:12069-74. [PMID: 16880389 PMCID: PMC1567698 DOI: 10.1073/pnas.0603866103] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
To date, it has not been possible to study antigen-specific T cell responses during primary infection of the genital tract. The low frequency of pathogen-specific T cells in a naïve mouse makes it difficult to monitor the initial events after antigen encounter. We developed a system to examine the response of pathogen-specific T cells in the genital mucosa after intrauterine infection. We identified the protective CD4(+) T cell antigen Cta1 from Chlamydia trachomatis and generated T cell receptor (TCR) transgenic (tg) mice with specificity for this protein. By transferring TCR tg T cells into naïve animals, we determined that Chlamydia-specific T cells were activated and proliferated in the lymph nodes draining the genital tract after primary intrauterine infection. Activated T cells migrated into the genital mucosa and secreted IFN-gamma. The development of Chlamydia-specific TCR tg mice provides an approach for dissecting how pathogen-specific T cells function in the genital tract.
Collapse
Affiliation(s)
- Nadia R. Roan
- Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, MA 02115
| | - Todd M. Gierahn
- Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, MA 02115
| | - Darren E. Higgins
- Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, MA 02115
| | - Michael N. Starnbach
- Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, MA 02115
- *To whom correspondence should be addressed. E-mail:
| |
Collapse
|