1
|
Pandey B, Wang Z, Jimenez A, Bhatia E, Jain R, Beach A, Maniar D, Hosten J, O'Farrell L, Vantucci C, Hur D, Noel R, Ringuist R, Smith C, Ochoa MA, Roy K. A multiadjuvant polysaccharide-amino acid-lipid (PAL) subunit nanovaccine generates robust systemic and lung-specific mucosal immune responses against SARS-CoV-2 in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.05.539395. [PMID: 37215018 PMCID: PMC10197586 DOI: 10.1101/2023.05.05.539395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Existing parenteral SARS-CoV-2 vaccines produce only limited mucosal responses, which are essential for reducing transmission and achieving sterilizing immunity. Appropriately designed mucosal boosters could overcome the shortcomings of parenteral vaccines and enhance pre- existing systemic immunity. Here we present a new protein subunit nanovaccine using multiadjuvanted (e.g. RIG-I: PUUC, TLR9: CpG) polysaccharide-amino acid-lipid nanoparticles (PAL-NPs) that can be delivered both intramuscularly (IM) and intranasally (IN) to generate balanced mucosal-systemic SARS-CoV-2 immunity. Mice receiving IM-Prime PUUC+CpG PAL- NPs, followed by an IN-Boost, developed high levels of IgA, IgG, and cellular immunity in the lung, and showed robust systemic humoral immunity. Interestingly, as a purely intranasal vaccine (IN-Prime/IN-Boost), PUUC+CpG PAL-NPs induced stronger lung-specific T cell immunity than IM-Prime/IN-Boost, and a comparable IgA and neutralizing antibodies, although with a lower systemic antibody response, indicating that a fully mucosal delivery route for SARS-CoV-2 vaccination may also be feasible. Our data suggest that PUUC+CpG PAL-NP subunit vaccine is a promising candidate for generating SARS-CoV-2 specific mucosal immunity.
Collapse
|
2
|
Bediaga NG, Garnham AL, Naselli G, Bandala-Sanchez E, Stone NL, Cobb J, Harbison JE, Wentworth JM, Ziegler AG, Couper JJ, Smyth GK, Harrison LC. Cytotoxicity-Related Gene Expression and Chromatin Accessibility Define a Subset of CD4+ T Cells That Mark Progression to Type 1 Diabetes. Diabetes 2022; 71:566-577. [PMID: 35007320 PMCID: PMC8893947 DOI: 10.2337/db21-0612] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 12/12/2021] [Indexed: 11/13/2022]
Abstract
Type 1 diabetes in children is heralded by a preclinical phase defined by circulating autoantibodies to pancreatic islet antigens. How islet autoimmunity is initiated and then progresses to clinical diabetes remains poorly understood. Only one study has reported gene expression in specific immune cells of children at risk associated with progression to islet autoimmunity. We analyzed gene expression with RNA sequencing in CD4+ and CD8+ T cells, natural killer (NK) cells, and B cells, and chromatin accessibility by assay for transposase-accessible chromatin sequencing (ATAC-seq) in CD4+ T cells, in five genetically at risk children with islet autoantibodies who progressed to diabetes over a median of 3 years ("progressors") compared with five children matched for sex, age, and HLA-DR who had not progressed ("nonprogressors"). In progressors, differentially expressed genes (DEGs) were largely confined to CD4+ T cells and enriched for cytotoxicity-related genes/pathways. Several top-ranked DEGs were validated in a semi-independent cohort of 13 progressors and 11 nonprogressors. Flow cytometry confirmed that progression was associated with expansion of CD4+ cells with a cytotoxic phenotype. By ATAC-seq, progression was associated with reconfiguration of regulatory chromatin regions in CD4+ cells, some linked to differentially expressed cytotoxicity-related genes. Our findings suggest that cytotoxic CD4+ T cells play a role in promoting progression to type 1 diabetes.
Collapse
Affiliation(s)
- Naiara G. Bediaga
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Alexandra L. Garnham
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Gaetano Naselli
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Esther Bandala-Sanchez
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
| | - Natalie L. Stone
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Joanna Cobb
- Murdoch Children’s Research Institute, Parkville, Australia
| | - Jessica E. Harbison
- Department of Endocrinology and Diabetes, Women’s and Children’s Hospital, North Adelaide, Australia
- Robinson Research Institute, The University of Adelaide, Adelaide, Australia
| | - John M. Wentworth
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
- Department of Diabetes and Endocrinology, Royal Melbourne Hospital, Parkville, Australia
| | - Annette-G. Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, Neuherberg, Germany
| | - Jennifer J. Couper
- Department of Endocrinology and Diabetes, Women’s and Children’s Hospital, North Adelaide, Australia
- Robinson Research Institute, The University of Adelaide, Adelaide, Australia
| | - Gordon K. Smyth
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- School of Mathematics and Statistics, The University of Melbourne, Parkville, Australia
| | - Leonard C. Harrison
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Australia
- Corresponding author: Leonard C. Harrison,
| |
Collapse
|
3
|
Perišić Nanut M, Pawelec G, Kos J. Human CD4+ T-Cell Clone Expansion Leads to the Expression of the Cysteine Peptidase Inhibitor Cystatin F. Int J Mol Sci 2021; 22:8408. [PMID: 34445118 PMCID: PMC8395124 DOI: 10.3390/ijms22168408] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/30/2021] [Accepted: 08/01/2021] [Indexed: 01/12/2023] Open
Abstract
The existence of CD4+ cytotoxic T cells (CTLs) at relatively high levels under different pathological conditions in vivo suggests their role in protective and/or pathogenic immune functions. CD4+ CTLs utilize the fundamental cytotoxic effector mechanisms also utilized by CD8+ CTLs and natural killer cells. During long-term cultivation, CD4+ T cells were also shown to acquire cytotoxic functions. In this study, CD4+ human T-cell clones derived from activated peripheral blood lymphocytes of healthy young adults were examined for the expression of cytotoxic machinery components. Cystatin F is a protein inhibitor of cysteine cathepsins, synthesized by CD8+ CTLs and natural killer cells. Cystatin F affects the cytotoxic efficacy of these cells by inhibiting the major progranzyme convertases cathepsins C and H as well as cathepsin L, which is involved in perforin activation. Here, we show that human CD4+ T-cell clones express the cysteine cathepsins that are involved in the activation of granzymes and perforin. CD4+ T-cell clones contained both the inactive, dimeric form as well as the active, monomeric form of cystatin F. As in CD8+ CTLs, cysteine cathepsins C and H were the major targets of cystatin F in CD4+ T-cell clones. Furthermore, CD4+ T-cell clones expressed the active forms of perforin and granzymes A and B. The levels of the cystatin F decreased with time in culture concomitantly with an increase in the activities of granzymes A and B. Therefore, our results suggest that cystatin F plays a role in regulating CD4+ T cell cytotoxicity. Since cystatin F can be secreted and taken up by bystander cells, our results suggest that CD4+ CTLs may also be involved in regulating immune responses through cystatin F secretion.
Collapse
Affiliation(s)
- Milica Perišić Nanut
- Department of Biotechnology, Jožef Stefan Institute, Jamova Cesta 39, 1000 Ljubljana, Slovenia;
| | - Graham Pawelec
- Interfaculty Institute for Cell Biology, Department of Immunology, University of Tübingen, Auf der Morgenstelle 15/3.008, 72076 Tübingen, Germany;
- Health Sciences North Research Institute, 56 Walford Rd, Sudbury, ON P3E 2H2, Canada
| | - Janko Kos
- Department of Biotechnology, Jožef Stefan Institute, Jamova Cesta 39, 1000 Ljubljana, Slovenia;
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva Cesta 7, 1000 Ljubljana, Slovenia
| |
Collapse
|
4
|
Cachot A, Bilous M, Liu YC, Li X, Saillard M, Cenerenti M, Rockinger GA, Wyss T, Guillaume P, Schmidt J, Genolet R, Ercolano G, Protti MP, Reith W, Ioannidou K, de Leval L, Trapani JA, Coukos G, Harari A, Speiser DE, Mathis A, Gfeller D, Altug H, Romero P, Jandus C. Tumor-specific cytolytic CD4 T cells mediate immunity against human cancer. SCIENCE ADVANCES 2021; 7:7/9/eabe3348. [PMID: 33637530 PMCID: PMC7909889 DOI: 10.1126/sciadv.abe3348] [Citation(s) in RCA: 189] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 01/14/2021] [Indexed: 05/04/2023]
Abstract
CD4 T cells have been implicated in cancer immunity for their helper functions. Moreover, their direct cytotoxic potential has been shown in some patients with cancer. Here, by mining single-cell RNA-seq datasets, we identified CD4 T cell clusters displaying cytotoxic phenotypes in different human cancers, resembling CD8 T cell profiles. Using the peptide-MHCII-multimer technology, we confirmed ex vivo the presence of cytolytic tumor-specific CD4 T cells. We performed an integrated phenotypic and functional characterization of these cells, down to the single-cell level, through a high-throughput nanobiochip consisting of massive arrays of picowells and machine learning. We demonstrated a direct, contact-, and granzyme-dependent cytotoxic activity against tumors, with delayed kinetics compared to classical cytotoxic lymphocytes. Last, we found that this cytotoxic activity was in part dependent on SLAMF7. Agonistic engagement of SLAMF7 enhanced cytotoxicity of tumor-specific CD4 T cells, suggesting that targeting these cells might prove synergistic with other cancer immunotherapies.
Collapse
Affiliation(s)
- Amélie Cachot
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Mariia Bilous
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, CH-1015, Switzerland
| | - Yen-Cheng Liu
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Xiaokang Li
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Margaux Saillard
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Mara Cenerenti
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, CH-1211, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, CH-1066, Switzerland
| | - Georg Alexander Rockinger
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Tania Wyss
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, CH-1015, Switzerland
| | - Philippe Guillaume
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Julien Schmidt
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Raphaël Genolet
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Giuseppe Ercolano
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, CH-1211, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, CH-1066, Switzerland
| | - Maria Pia Protti
- Tumor Immunology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milan, Italy
| | - Walter Reith
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, CH-1211, Switzerland
| | - Kalliopi Ioannidou
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, CH-1011, Switzerland
| | - Laurence de Leval
- Institute of Pathology, Department of Laboratory Medicine and Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, CH-1011, Switzerland
| | - Joseph A Trapani
- Peter MacCallum Cancer Centre, 305 Grattan Street, Melbourne 3000, Australia
| | - George Coukos
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Alexandre Harari
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Daniel E Speiser
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Alexander Mathis
- Harvard University, Cambridge, MA, USA
- Center for Neuroprosthetics, Center for Intelligent Systems, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
- Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, CH-1015, Switzerland
| | - David Gfeller
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, CH-1015, Switzerland
| | - Hatice Altug
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, CH-1015, Switzerland
| | - Pedro Romero
- Department of Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, CH-1066, Switzerland
| | - Camilla Jandus
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, CH-1211, Switzerland.
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, CH-1066, Switzerland
| |
Collapse
|
5
|
Pérez-Galarza J, Prócel C, Cañadas C, Aguirre D, Pibaque R, Bedón R, Sempértegui F, Drexhage H, Baldeón L. Immune Response to SARS-CoV-2 Infection in Obesity and T2D: Literature Review. Vaccines (Basel) 2021; 9:102. [PMID: 33572702 PMCID: PMC7911386 DOI: 10.3390/vaccines9020102] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/09/2021] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
In December 2019, a novel coronavirus known as SARS-CoV-2 was first detected in Wuhan, China, causing outbreaks of the coronavirus disease COVID-19 that has now spread globally. For this reason, The World Health Organization (WHO) declared COVID-19 a public health emergency in March 2020. People living with pre-existing conditions such as obesity, cardiovascular diseases, type 2 diabetes (T2D), and chronic kidney and lung diseases, are prone to develop severe forms of disease with fatal outcomes. Metabolic diseases such as obesity and T2D alter the balance of innate and adaptive responses. Both diseases share common features characterized by augmented adiposity associated with a chronic systemic low-grade inflammation, senescence, immunoglobulin glycation, and abnormalities in the number and function of adaptive immune cells. In obese and T2D patients infected by SARS-CoV-2, where immune cells are already hampered, this response appears to be stronger. In this review, we describe the abnormalities of the immune system, and summarize clinical findings of COVID-19 patients with pre-existing conditions such as obesity and T2D as this group is at greater risk of suffering severe and fatal clinical outcomes.
Collapse
Affiliation(s)
- Jorge Pérez-Galarza
- Research Institute of Biomedicine, Central University of Ecuador, Quito 170201, Ecuador; (J.P.-G.); (C.C.); (D.A.); (R.P.)
- Faculty of Medicine, Central University of Ecuador, Quito 170403, Ecuador; (R.B.); (F.S.)
| | | | - Cristina Cañadas
- Research Institute of Biomedicine, Central University of Ecuador, Quito 170201, Ecuador; (J.P.-G.); (C.C.); (D.A.); (R.P.)
| | - Diana Aguirre
- Research Institute of Biomedicine, Central University of Ecuador, Quito 170201, Ecuador; (J.P.-G.); (C.C.); (D.A.); (R.P.)
| | - Ronny Pibaque
- Research Institute of Biomedicine, Central University of Ecuador, Quito 170201, Ecuador; (J.P.-G.); (C.C.); (D.A.); (R.P.)
| | - Ricardo Bedón
- Faculty of Medicine, Central University of Ecuador, Quito 170403, Ecuador; (R.B.); (F.S.)
- Hospital General Docente de Calderón, Quito 170201, Ecuador
| | - Fernando Sempértegui
- Faculty of Medicine, Central University of Ecuador, Quito 170403, Ecuador; (R.B.); (F.S.)
| | - Hemmo Drexhage
- Immunology Department, Erasmus Medical Center, 3015 Rotterdam, The Netherlands;
| | - Lucy Baldeón
- Research Institute of Biomedicine, Central University of Ecuador, Quito 170201, Ecuador; (J.P.-G.); (C.C.); (D.A.); (R.P.)
- Faculty of Medicine, Central University of Ecuador, Quito 170403, Ecuador; (R.B.); (F.S.)
| |
Collapse
|
6
|
Law JC, Koh WH, Budylowski P, Lin J, Yue F, Abe KT, Rathod B, Girard M, Li Z, Rini JM, Mubareka S, McGeer A, Chan AK, Gingras AC, Watts TH, Ostrowski MA. Systematic Examination of Antigen-Specific Recall T Cell Responses to SARS-CoV-2 versus Influenza Virus Reveals a Distinct Inflammatory Profile. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 206:37-50. [PMID: 33208459 PMCID: PMC7750861 DOI: 10.4049/jimmunol.2001067] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/02/2020] [Indexed: 12/15/2022]
Abstract
There is a pressing need for an in-depth understanding of immunity to SARS-CoV-2. In this study, we investigated human T cell recall responses to fully glycosylated spike trimer, recombinant N protein, as well as to S, N, M, and E peptide pools in the early convalescent phase and compared them with influenza-specific memory responses from the same donors. All subjects showed SARS-CoV-2-specific T cell responses to at least one Ag. Both SARS-CoV-2-specific and influenza-specific CD4+ T cell responses were predominantly of the central memory phenotype; however SARS-CoV-2-specific CD4+ T cells exhibited a lower IFN-γ to TNF ratio compared with influenza-specific memory responses from the same donors, independent of disease severity. SARS-CoV-2-specific T cells were less multifunctional than influenza-specific T cells, particularly in severe cases, potentially suggesting exhaustion. Most SARS-CoV-2-convalescent subjects also produced IFN-γ in response to seasonal OC43 S protein. We observed granzyme B+/IFN-γ+, CD4+, and CD8+ proliferative responses to peptide pools in most individuals, with CD4+ T cell responses predominating over CD8+ T cell responses. Peripheral T follicular helper (pTfh) responses to S or N strongly correlated with serum neutralization assays as well as receptor binding domain-specific IgA; however, the frequency of pTfh responses to SARS-CoV-2 was lower than the frequency of pTfh responses to influenza virus. Overall, T cell responses to SARS-CoV-2 are robust; however, CD4+ Th1 responses predominate over CD8+ T cell responses, have a more inflammatory profile, and have a weaker pTfh response than the response to influenza virus within the same donors, potentially contributing to COVID-19 disease.
Collapse
Affiliation(s)
- Jaclyn C Law
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Wan Hon Koh
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario M5S 3H2, Canada
| | - Patrick Budylowski
- Department of Medicine, University of Toronto, Toronto, Ontario M5S 3H2, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Jonah Lin
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - FengYun Yue
- Department of Medicine, University of Toronto, Toronto, Ontario M5S 3H2, Canada
| | - Kento T Abe
- Lunenfeld-Tanenbaum Research Institute at Mt. Sinai Hospital, Sinai Health System, Toronto, Ontario M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Bhavisha Rathod
- Lunenfeld-Tanenbaum Research Institute at Mt. Sinai Hospital, Sinai Health System, Toronto, Ontario M5G 1X5, Canada
| | - Melanie Girard
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Zhijie Li
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - James M Rini
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Samira Mubareka
- Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Allison McGeer
- Lunenfeld-Tanenbaum Research Institute at Mt. Sinai Hospital, Sinai Health System, Toronto, Ontario M5G 1X5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Adrienne K Chan
- Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
- Division of Infectious Diseases, Department of Medicine, University of Toronto, Toronto, Ontario M5S 3H2, Canada; and
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute at Mt. Sinai Hospital, Sinai Health System, Toronto, Ontario M5G 1X5, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Tania H Watts
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada;
| | - Mario A Ostrowski
- Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario M5S 3H2, Canada
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario M5B 1W8, Canada
| |
Collapse
|
7
|
Rao G, Latha K, Ott M, Sabbagh A, Marisetty A, Ling X, Zamler D, Doucette TA, Yang Y, Kong LY, Wei J, Fuller GN, Benavides F, Sonabend AM, Long J, Li S, Curran M, Heimberger AB. Anti-PD-1 Induces M1 Polarization in the Glioma Microenvironment and Exerts Therapeutic Efficacy in the Absence of CD8 Cytotoxic T Cells. Clin Cancer Res 2020; 26:4699-4712. [PMID: 32554515 DOI: 10.1158/1078-0432.ccr-19-4110] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/16/2020] [Accepted: 06/11/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE Anti-programmed cell death protein 1 (PD-1) therapy has demonstrated inconsistent therapeutic results in patients with glioblastoma (GBM) including those with profound impairments in CD8 T-cell effector responses. EXPERIMENTAL DESIGN We ablated the CD8α gene in BL6 mice and intercrossed them with Ntv-a mice to determine how CD8 T cells affect malignant progression in forming endogenous gliomas. Tumor-bearing mice were treated with PD-1 to determine the efficacy of this treatment in the absence of T cells. The tumor microenvironment of treated and control mice was analyzed by IHC and FACS. RESULTS We observed a survival benefit in immunocompetent mice with endogenously arising intracranial glioblastomas after intravenous administration of anti-PD-1. The therapeutic effect of PD-1 administration persisted in mice even after genetic ablation of the CD8 gene (CD8-/-). CD11b+ and Iba1+ monocytes and macrophages were enriched in the glioma microenvironment of the CD8-/- mice. The macrophages and microglia assumed a proinflammatory M1 response signature in the setting of anti-PD-1 blockade through the elimination of PD-1-expressing macrophages and microglia in the tumor microenvironment. Anti-PD-1 can inhibit the proliferation of and induce apoptosis of microglia through antibody-dependent cellular cytotoxicity, as fluorescently labeled anti-PD-1 was shown to gain direct access to the glioma microenvironment. CONCLUSIONS Our results show that the therapeutic effect of anti-PD-1 blockade in GBM may be mediated by the innate immune system, rather than by CD8 T cells. Anti-PD-1 immunologically modulates innate immunity in the glioma microenvironment-likely a key mode of activity.
Collapse
Affiliation(s)
- Ganesh Rao
- Department of Neurosurgery, Baylor College of Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Khatri Latha
- Department of Neurosurgery, Baylor College of Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Martina Ott
- Department of Neurosurgery, Baylor College of Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aria Sabbagh
- Department of Neurosurgery, Baylor College of Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anantha Marisetty
- Department of Neurosurgery, Baylor College of Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiaoyang Ling
- Department of Neurosurgery, Baylor College of Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Daniel Zamler
- Department of Genomic Medicine and Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tiffany A Doucette
- Department of Neurosurgery, Baylor College of Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yuhui Yang
- Department of Neurosurgery, Baylor College of Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ling-Yuan Kong
- Department of Neurosurgery, Baylor College of Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jun Wei
- Department of Neurosurgery, Baylor College of Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gregory N Fuller
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Fernando Benavides
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Adam M Sonabend
- Department of Neurosurgery, Feinberg School of Medicine, Robert H Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois
| | - James Long
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shulin Li
- Department of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael Curran
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Amy B Heimberger
- Department of Neurosurgery, Baylor College of Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
8
|
Sanchez-Martinez A, Perdomo-Celis F, Acevedo-Saenz L, Rugeles MT, Velilla PA. Cytotoxic CD4 + T-cells during HIV infection: Targets or weapons? J Clin Virol 2019; 119:17-23. [PMID: 31445411 DOI: 10.1016/j.jcv.2019.08.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 07/16/2019] [Accepted: 08/13/2019] [Indexed: 12/23/2022]
Abstract
Classically, CD4+ T-cells have been referred as cytokine-producing cells and important players in immune responses by providing soluble factors that potentiate several effector immune functions. However, it is now evident that CD4+ T-cells can also elaborate cytotoxic responses, inducing apoptosis of target cells. Cytotoxic CD4+ T cells (CD4+ CTLs), exhibit cytolytic functions that resemble those of CD8+ T-cells; in fact, there is evidence suggesting that they may have a role in the control of viral infections. In this article, we discuss the role of CD4+ CTLs during HIV infection, where CD4+ CTLs have been associated with viral control and slow disease progression. In addition, we address the implication of CD4+ CTLs in the context of antiretroviral therapy and the partial reconstitution of CD8+ T-cells effector function.
Collapse
Affiliation(s)
| | - Federico Perdomo-Celis
- Grupo Inmunovirologia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | - Liliana Acevedo-Saenz
- Grupo Inmunovirologia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia; Grupo de Investigación Enfermería-CES, Facultad de Enfermería, Universidad CES, Medellin, Colombia
| | - Maria T Rugeles
- Grupo Inmunovirologia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia
| | - Paula A Velilla
- Grupo Inmunovirologia, Facultad de Medicina, Universidad de Antioquia, Medellin, Colombia.
| |
Collapse
|
9
|
Phetsouphanh C, Aldridge D, Marchi E, Munier CML, Meyerowitz J, Murray L, Van Vuuren C, Goedhals D, Fidler S, Kelleher A, Klenerman P, Frater J. Maintenance of Functional CD57+ Cytolytic CD4+ T Cells in HIV+ Elite Controllers. Front Immunol 2019; 10:1844. [PMID: 31440240 PMCID: PMC6694780 DOI: 10.3389/fimmu.2019.01844] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 07/22/2019] [Indexed: 11/29/2022] Open
Abstract
Cytolytic CD4+ T cells play a prominent role in chronic viral infection. CD4+ CTLs clones specific for HIV-1 Nef and Gag are capable of killing HIV-1 infected CD4+ T cells and macrophages. Additionally, HIV-specific cytolytic CD4+ T cell responses in acute HIV infection are predictive of disease progression. CD57 expression on CD4s identifies cytolytic cells. These cells were dramatically increased in chronic HIV infection. CD57 expression correlated with cytolytic granules, granzyme B and perforin expression. They express lower CCR5 compared to CD57- cells, have less HIV total DNA, and were a minor component of the HIV reservoir. A small percentage of CD57+ CD4+ CTLs from EC were HIV-specific, could upregulate IFNγ with Gag peptide stimulation, express cytolytic granule markers and maintain TbethighEomes+ transcription factor phenotype. This was not observed in viraemic controllers. The maintenance of HIV-specific CD4 cytolytic function in Elite controllers together with CD8 CTLs may be important for the control of HIV viraemia and of potential relevance to cure strategies.
Collapse
Affiliation(s)
| | - Daniel Aldridge
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Emanuele Marchi
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - C. Mee Ling Munier
- Department of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Jodi Meyerowitz
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Lyle Murray
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | | | - Dominique Goedhals
- National Health Laboratory Service, Division of Virology, University of the Free State, Bloemfontein, South Africa
| | | | - Anthony Kelleher
- Department of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - John Frater
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
10
|
Abstract
Chronic lymphocytic leukaemia (CLL) has long been thought to be an immunosuppressive disease and abnormalities in T-cell subset distribution and function have been observed in many studies. However, the role of T cells (if any) in disease progression remains unclear and has not been directly studied. This has changed with the advent of new therapies, such as chimeric antigen receptor-T cells, which actively use retargeted patient-derived T cells as "living drugs" for CLL. However complete responses are relatively low (~26%) and recent studies have suggested the differentiation status of patient T cells before therapy may influence efficacy. Non-chemotherapeutic drugs, such as idelalisib and ibrutinib, also have an impact on T cell populations in CLL patients. This review will highlight what is known about T cells in CLL during disease progression and after treatment, and discuss the prospects of using T cells as predictive biomarkers for immune status and response to therapy.
Collapse
MESH Headings
- Adenine/analogs & derivatives
- B-Lymphocytes/immunology
- B-Lymphocytes/pathology
- Humans
- Immunotherapy, Adoptive
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Piperidines
- Purines/therapeutic use
- Pyrazoles/therapeutic use
- Pyrimidines/therapeutic use
- Quinazolinones/therapeutic use
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/pathology
Collapse
Affiliation(s)
- Stephen Man
- Section of Haematology, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, UK
| | - Peter Henley
- Section of Haematology, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, UK
| |
Collapse
|
11
|
Lyadova I, Nikitina I. Cell Differentiation Degree as a Factor Determining the Role for Different T-Helper Populations in Tuberculosis Protection. Front Immunol 2019; 10:972. [PMID: 31134070 PMCID: PMC6517507 DOI: 10.3389/fimmu.2019.00972] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 04/16/2019] [Indexed: 12/15/2022] Open
Abstract
Efficient tuberculosis (TB) control depends on early TB prediction and prevention. Solution to these tasks requires knowledge of TB protection correlates (TB CoPs), i.e., laboratory markers that are mechanistically involved in the protection and which allow to determine how well an individual is protected against TB or how efficient the candidate TB vaccine is. The search for TB CoPs has been largely focused on different T-helper populations, however, the data are controversial, and no reliable CoPs are still known. Here we discuss the role of different T-helper populations in TB protection focusing predominantly on Th17, “non-classical” Th1 (Th1*) and “classical” Th1 (cTh1) populations. We analyze how these populations differ besides their effector activity and suggest the hypothesis that: (i) links the protective potential of Th17, Th1*, and cTh1 to their differentiation degree and plasticity; (ii) implies different roles of these populations in response to vaccination, latent TB infection (LTBI), and active TB. One of the clinically relevant outcomes of this hypothesis is that over-stimulating T cells during vaccination and biasing T cell response toward the preferential generation of Th1 are not beneficial. The review sheds new light on the problem of TB CoPs and will help develop better strategies for TB control.
Collapse
Affiliation(s)
- Irina Lyadova
- Laboratory of Cellular and Molecular Mechanisms of Histogenesis, Koltsov Institute of Developmental Biology, Moscow, Russia.,Laboratory of Biotechnology, Department of Immunology, Central Tuberculosis Research Institute, Moscow, Russia
| | - Irina Nikitina
- Laboratory of Cellular and Molecular Mechanisms of Histogenesis, Koltsov Institute of Developmental Biology, Moscow, Russia.,Laboratory of Biotechnology, Department of Immunology, Central Tuberculosis Research Institute, Moscow, Russia
| |
Collapse
|
12
|
Mattoo H, Stone JH, Pillai S. Clonally expanded cytotoxic CD4 + T cells and the pathogenesis of IgG4-related disease. Autoimmunity 2017; 50:19-24. [PMID: 28166682 DOI: 10.1080/08916934.2017.1280029] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
IgG4-related disease (IgG4-RD) is a systemic condition of unknown cause characterized by highly fibrotic lesions, with dense lymphoplasmacytic infiltrates containing a preponderance of IgG4-expressing plasma cells. CD4+ T cells and B cells constitute the major inflammatory cell populations in IgG4-RD lesions. IgG4-RD patients with active, untreated disease show a marked expansion of plasmablasts in the circulation. Although the therapeutic depletion of B cells suggests a role for these cells in the disease, a direct role for B cells or IgG4 in the pathogenesis of IgG4-RD is yet to be demonstrated. Among the CD4+ T-cell subsets, Th2 cells were initially thought to contribute to IgG4-RD pathogenesis, but many previous studies were confounded by the concomitant history of allergic diseases in the patients studied and the failure to use multi-color staining to definitively identify T-cell subsets in tissue samples. More recently, using an unbiased approach to characterize CD4+ T-cell subsets in patients with IgG4-RD - based on their clonal expansion and ability to infiltrate affected tissue sites - CD4+ CTLs have been identified as the major CD4+ T-cell subset in disease lesions as well as in the circulation. CD4+ CTLs in affected tissues secrete pro-fibrotic cytokines including IL-1β, TGF-β1, and IFN-γ as well as cytolytic molecules such as perforin and granzymes A and B. In this review, we examine possible mechanisms by which activated B cells and plasmablasts may collaborate with the expanded CD4+ CTLs in driving the fibrotic pathology of the disease and describe the lacunae in the field and in our understanding of IgG4-RD pathogenesis.
Collapse
Affiliation(s)
- Hamid Mattoo
- a Massachusetts General Hospital, Harvard Medical School , Boston , MA , USA
| | - John H Stone
- a Massachusetts General Hospital, Harvard Medical School , Boston , MA , USA
| | - Shiv Pillai
- a Massachusetts General Hospital, Harvard Medical School , Boston , MA , USA
| |
Collapse
|
13
|
Phetsouphanh C, Pillai S, Zaunders JJ. Editorial: Cytotoxic CD4+ T Cells in Viral Infections. Front Immunol 2017; 8:1729. [PMID: 29255471 PMCID: PMC5723143 DOI: 10.3389/fimmu.2017.01729] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 11/22/2017] [Indexed: 11/13/2022] Open
Affiliation(s)
- Chansavath Phetsouphanh
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Shiv Pillai
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - John J Zaunders
- The Kirby Institute, University of New South Wales, Sydney, NSW, Australia.,St Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Sydney, NSW, Australia
| |
Collapse
|
14
|
Kim HY, Yoo TH, Hwang Y, Lee GH, Kim B, Jang J, Yu HT, Kim MC, Cho JY, Lee CJ, Kim HC, Park S, Lee WW. Indoxyl sulfate (IS)-mediated immune dysfunction provokes endothelial damage in patients with end-stage renal disease (ESRD). Sci Rep 2017; 7:3057. [PMID: 28596556 PMCID: PMC5465082 DOI: 10.1038/s41598-017-03130-z] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 04/25/2017] [Indexed: 12/19/2022] Open
Abstract
Progressive renal failure causes uremia-related immune dysfunction, which features a chronic inflammatory milieu. Given the central role of end-stage renal disease (ESRD)-related immune dysfunction in the pathogenesis of cardiovascular diseases (CVDs), much attention has been focused on how uremic toxins affect cellular immunity and the mechanisms underlying pathogenesis of atherosclerosis in ESRD patients. Here, we investigated the characteristics of monocytes and CD4+ T cells in ESRD patients and the immune responses induced by indoxyl sulfate (IS), a key uremic toxin, in order to explore the pathogenic effects of these cells on vascular endothelial cells. In ESRD patients, monocytes respond to IS through the aryl hydrocarbon receptor (AhR) and consequently produce increased levels of TNF-α. Upon stimulation with TNF-α, human vascular endothelial cells produce copious amounts of CX3CL1, a chemokine ligand of CX3CR1 that is highly expressed on CD4+CD28-T cells, the predominantly expanded cell type in ESRD patients. A migration assay showed that CD4+CD28- T cells were preferentially recruited by CX3CL1. Moreover, activated CD4+CD28- T cells exhibited cytotoxic capability allowing for the induction of apoptosis in HUVECs. Our findings suggest that in ESRD, IS-mediated immune dysfunction may cause vascular endothelial cell damage and thus, this toxin plays a pivotal role in the pathogenesis of CVD.
Collapse
Affiliation(s)
- Hee Young Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea
| | - Tae-Hyun Yoo
- Division of Nephrology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Yuri Hwang
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea
- BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea
| | - Ga Hye Lee
- BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Bonah Kim
- BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Jiyeon Jang
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea
| | - Hee Tae Yu
- Division of Cardiology, Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Min Chang Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, South Korea
| | - Joo-Youn Cho
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, South Korea
| | - Chan Joo Lee
- Department of Health Promotion and Disease Prevention, Severance Hospital, Seoul, South Korea
| | - Hyeon Chang Kim
- Cardiovascular and Metabolic Diseases Etiology Research Center and Department of Preventive Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Sungha Park
- Division of Cardiology, Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, South Korea.
| | - Won-Woo Lee
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea.
- BK21Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul, South Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.
- Ischemic/Hypoxic Disease Institute and Institute of Infectious Diseases, Seoul National University College of Medicine; Seoul National University Hospital Biomedical Research Institute, Seoul, South Korea.
| |
Collapse
|
15
|
Rieckmann JC, Geiger R, Hornburg D, Wolf T, Kveler K, Jarrossay D, Sallusto F, Shen-Orr SS, Lanzavecchia A, Mann M, Meissner F. Social network architecture of human immune cells unveiled by quantitative proteomics. Nat Immunol 2017; 18:583-593. [DOI: 10.1038/ni.3693] [Citation(s) in RCA: 229] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 01/26/2017] [Indexed: 02/08/2023]
|
16
|
Muraro E, Merlo A, Martorelli D, Cangemi M, Dalla Santa S, Dolcetti R, Rosato A. Fighting Viral Infections and Virus-Driven Tumors with Cytotoxic CD4 + T Cells. Front Immunol 2017; 8:197. [PMID: 28289418 PMCID: PMC5327441 DOI: 10.3389/fimmu.2017.00197] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/09/2017] [Indexed: 12/18/2022] Open
Abstract
CD4+ T cells have been and are still largely regarded as the orchestrators of immune responses, being able to differentiate into distinct T helper cell populations based on differentiation signals, transcription factor expression, cytokine secretion, and specific functions. Nonetheless, a growing body of evidence indicates that CD4+ T cells can also exert a direct effector activity, which depends on intrinsic cytotoxic properties acquired and carried out along with the evolution of several pathogenic infections. The relevant role of CD4+ T cell lytic features in the control of such infectious conditions also leads to their exploitation as a new immunotherapeutic approach. This review aims at summarizing currently available data about functional and therapeutic relevance of cytotoxic CD4+ T cells in the context of viral infections and virus-driven tumors.
Collapse
Affiliation(s)
- Elena Muraro
- Immunopathology and Cancer Biomarkers, Traslational Research Department, IRCCS, C.R.O. National Cancer Institute, Aviano, Pordenone, Italy
| | - Anna Merlo
- Department of Immunology and Blood Transfusions, San Bortolo Hospital, Vicenza, Italy
| | - Debora Martorelli
- Immunopathology and Cancer Biomarkers, Traslational Research Department, IRCCS, C.R.O. National Cancer Institute, Aviano, Pordenone, Italy
| | - Michela Cangemi
- Immunopathology and Cancer Biomarkers, Traslational Research Department, IRCCS, C.R.O. National Cancer Institute, Aviano, Pordenone, Italy
| | | | - Riccardo Dolcetti
- Immunopathology and Cancer Biomarkers, Traslational Research Department, IRCCS, C.R.O. National Cancer Institute, Aviano, Pordenone, Italy
- Translational Research Institute, University of Queensland Diamantina Institute, Brisbane, QLD, Australia
| | - Antonio Rosato
- Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, Oncology and Immunology Section, University of Padova, Padova, Italy
| |
Collapse
|
17
|
Juno JA, van Bockel D, Kent SJ, Kelleher AD, Zaunders JJ, Munier CML. Cytotoxic CD4 T Cells-Friend or Foe during Viral Infection? Front Immunol 2017; 8:19. [PMID: 28167943 PMCID: PMC5253382 DOI: 10.3389/fimmu.2017.00019] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 01/05/2017] [Indexed: 01/03/2023] Open
Abstract
CD4 T cells with cytotoxic function were once thought to be an artifact due to long-term in vitro cultures but have in more recent years become accepted and reported in the literature in response to a number of viral infections. In this review, we focus on cytotoxic CD4 T cells in the context of human viral infections and in some infections that affect mice and non-human primates. We examine the effector mechanisms used by cytotoxic CD4 cells, the phenotypes that describe this population, and the transcription factors and pathways that lead to their induction following infection. We further consider the cells that are the predominant targets of this effector subset and describe the viral infections in which CD4 cytotoxic T lymphocytes have been shown to play a protective or pathologic role. Cytotoxic CD4 T cells are detected in the circulation at much higher levels than previously realized and are now recognized to have an important role in the immune response to viral infections.
Collapse
Affiliation(s)
- Jennifer A Juno
- Department of Microbiology and Immunology, Peter Doherty Institute, University of Melbourne , Melbourne, VIC , Australia
| | - David van Bockel
- Immunovirology and Pathogenesis Program, The Kirby Institute for Infection and Immunity in Society, University of New South Wales Australia , Sydney, NSW , Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, Peter Doherty Institute, University of Melbourne, Melbourne, VIC, Australia; Melbourne Sexual Health Centre, Department of Infectious Diseases, Alfred Health, Central Clinical School, Monash University, Melbourne, VIC, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Parkville, VIC, Australia
| | - Anthony D Kelleher
- Immunovirology and Pathogenesis Program, The Kirby Institute for Infection and Immunity in Society, University of New South Wales Australia, Sydney, NSW, Australia; St Vincent's Hospital, Sydney, NSW, Australia
| | - John J Zaunders
- Immunovirology and Pathogenesis Program, The Kirby Institute for Infection and Immunity in Society, University of New South Wales Australia, Sydney, NSW, Australia; St Vincent's Hospital, Sydney, NSW, Australia
| | - C Mee Ling Munier
- Immunovirology and Pathogenesis Program, The Kirby Institute for Infection and Immunity in Society, University of New South Wales Australia , Sydney, NSW , Australia
| |
Collapse
|
18
|
Escarra-Senmarti M, Bueno-Topete MR, Jave-Suarez LF, Gomez-Bañuelos E, Gutierrez-Franco J, Vega-Magaña N, Aguilar-Lemarroy A, Pereira-Suarez AL, Haramati J, Del Toro-Arreola S. Loss of CD28 within CD4 + T cell subsets from cervical cancer patients is accompanied by the acquisition of intracellular perforin, and is further enhanced by NKG2D expression. Immunol Lett 2017; 182:30-38. [PMID: 28087292 DOI: 10.1016/j.imlet.2017.01.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 01/06/2017] [Accepted: 01/09/2017] [Indexed: 10/20/2022]
Abstract
CD28 is well characterized as an essential co-stimulatory receptor critical for activation, proliferation and survival processes in CD4+ T cells. Populations of CD4+CD28null T cells, with apparently contradictory physiological roles, have recently been reported, along with the co-expression of the NK activating receptor NKG2D, in autoimmune diseases and chronic viral inflammation. Paradoxically, studies in cancer suggest that an expanded CD4+NKG2D+ population may be armed with immunosuppressive properties. We have recently reported the existence of two separate CD4+NKG2D+ T cell populations, which were defined by the presence or absence of the co-stimulatory molecule CD28, with the CD4+CD28nullNKG2D+ population more frequently observed in women with cervical cancer. This has led to the present effort to further characterize this population and to determine if the loss of CD28 influences the acquisition of cytotoxic or regulatory markers. In the present work, a multicolor flow cytometry protocol was used to analyze the expression of cytotoxic and immunoregulatory markers on circulating CD4+ T cells characterized by the presence or absence of CD28 and NKG2D in patients with invasive cervical carcinoma and age/gender-matched healthy controls. A noticeable expansion of CD4+CD28null cells, many of them NKG2D+, were observed in selected cervical cancer samples. This CD4+CD28null T cell population was characterized by a lack of immunoregulatory markers, as well as very low basal levels of intracellular IFN-γ, TNF-α, TGF-β, and IL-10. Intracellular perforin, however, was found to be significantly increased in this CD4+CD28null population, and increases in the mean fluorescence intensity of perforin were found to be enhanced by the presence of NKG2D. In conclusion, our data provide the first evidence of a strict link between the absence of CD28 and the expression of perforin, which is likewise enhanced by the expression of NKG2D, within selected CD4+ T cells from cervical cancer patients.
Collapse
Affiliation(s)
- Marta Escarra-Senmarti
- Instituto de Enfermedades Crónico-Degenerativas, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico.
| | - Miriam Ruth Bueno-Topete
- Instituto de Enfermedades Crónico-Degenerativas, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico.
| | - Luis Felipe Jave-Suarez
- División de Inmunología, CIBO, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, Mexico.
| | - Eduardo Gomez-Bañuelos
- Instituto de Investigación en Reumatología y del Sistema Músculo Esquelético, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico.
| | - Jorge Gutierrez-Franco
- Instituto de Enfermedades Crónico-Degenerativas, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico.
| | - Natali Vega-Magaña
- Instituto de Enfermedades Crónico-Degenerativas, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico.
| | - Adriana Aguilar-Lemarroy
- División de Inmunología, CIBO, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, Mexico.
| | - Ana Laura Pereira-Suarez
- Laboratorio de Inmunología, Departamento de Fisiología, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico.
| | - Jesse Haramati
- Laboratorio de Inmunobiología, Departamento de Biología Celular y Molecular, CUCBA, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico.
| | - Susana Del Toro-Arreola
- Instituto de Enfermedades Crónico-Degenerativas, Departamento de Biología Molecular y Genómica, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico; Laboratorio de Inmunología, Departamento de Fisiología, CUCS, Universidad de Guadalajara, Guadalajara, Jalisco, Mexico.
| |
Collapse
|
19
|
Park Y, Moon SJ, Lee SW. Lineage re-commitment of CD4CD8αα intraepithelial lymphocytes in the gut. BMB Rep 2016; 49:11-7. [PMID: 26592937 PMCID: PMC4914207 DOI: 10.5483/bmbrep.2016.49.1.242] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Indexed: 12/28/2022] Open
Abstract
The gastrointestinal tract forms the largest surface in our body with constantly
being exposed to various antigens, which provides unique microenvironment for
the immune system in the intestine. Accordingly, the gut epithelium harbors the
most T lymphocytes in the body as intraepithelial
lymphocytes (IELs), which are phenotypically and
functionally heterogeneous populations, distinct from the conventional mature T
cells in the periphery. IELs arise either from pre-committed thymic precursors
(natural IELs) or from conventional CD4 or CD8αβ T cells in response
to peripheral antigens (induced IELs), both of which commonly express CD8α
homodimers (CD8αα). Although lineage commitment to either conventional
CD4 T helper (Th) or cytotoxic CD8αβ T cells as well as their
respective co-receptor expression are mutually exclusive and irreversible
process, CD4 T cells can be redirected to the CD8 IELs with high cytolytic
activity upon migration to the gut epithelium. Recent reports show that master
transcription factors for CD4 and CD8 T cells, ThPOK (Th-inducing
BTB/POZ-Kruppel-like factor) and Runx3 (Runt related transcription factor 3),
respectively, are the key regulators for re-programming of CD4 T cells to CD8
lineage in the intestinal epithelium. This review will focus on the unique
differentiation process of IELs, particularly lineage re-commitment of CD4 IELs.
[BMB Reports 2016; 49(1): 11-17]
Collapse
Affiliation(s)
- Yunji Park
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Sook-Jin Moon
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Seung-Woo Lee
- Division of Integrative Biosciences and Biotechnology and Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| |
Collapse
|
20
|
Malignant melanoma—The cradle of anti-neoplastic immunotherapy. Crit Rev Oncol Hematol 2016; 106:25-54. [DOI: 10.1016/j.critrevonc.2016.04.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 03/14/2016] [Accepted: 04/25/2016] [Indexed: 02/07/2023] Open
|
21
|
Burel JG, Apte SH, Groves PL, Klein K, McCarthy JS, Doolan DL. Reduced Plasmodium Parasite Burden Associates with CD38+ CD4+ T Cells Displaying Cytolytic Potential and Impaired IFN-γ Production. PLoS Pathog 2016; 12:e1005839. [PMID: 27662621 PMCID: PMC5035011 DOI: 10.1371/journal.ppat.1005839] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 08/02/2016] [Indexed: 11/19/2022] Open
Abstract
Using a unique resource of samples from a controlled human malaria infection (CHMI) study, we identified a novel population of CD4+ T cells whose frequency in the peripheral blood was inversely correlated with parasite burden following P. falciparum infection. These CD4+ T cells expressed the multifunctional ectoenzyme CD38 and had unique features that distinguished them from other CD4+ T cells. Specifically, their phenotype was associated with proliferation, activation and cytotoxic potential as well as significantly impaired production of IFN-γ and other cytokines and reduced basal levels of activated STAT1. A CD38+ CD4+ T cell population with similar features was identified in healthy uninfected individuals, at lower frequency. CD38+ CD4+ T cells could be generated in vitro from CD38- CD4+ T cells after antigenic or mitogenic stimulation. This is the first report of a population of CD38+ CD4+ T cells with a cytotoxic phenotype and markedly impaired IFN-γ capacity in humans. The expansion of this CD38+ CD4+ T population following infection and its significant association with reduced blood-stage parasite burden is consistent with an important functional role for these cells in protective immunity to malaria in humans. Their ubiquitous presence in humans suggests that they may have a broad role in host-pathogen defense. TRIAL REGISTRATION ClinicalTrials.gov clinical trial numbers ACTRN12612000814875, ACTRN12613000565741 and ACTRN12613001040752.
Collapse
Affiliation(s)
- Julie G. Burel
- Molecular Vaccinology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
- The University of Queensland, School of Medicine, Brisbane, Australia
| | - Simon H. Apte
- Molecular Vaccinology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Penny L. Groves
- Molecular Vaccinology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Kerenaftali Klein
- Statistics Unit, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - James S. McCarthy
- Clinical Tropical Medicine Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Denise L. Doolan
- Molecular Vaccinology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia
- The University of Queensland, School of Medicine, Brisbane, Australia
- Centre for Biosecurity and Tropical Infectious Diseases, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
- * E-mail:
| |
Collapse
|
22
|
Weng J, Baio FE, Moriarty KE, Torikai H, Wang H, Liu Z, Maiti SN, Gwak D, Popescu MS, Cha SC, Cooper LJN, Neelapu SS, Kwak LW. Targeting B-cell malignancies through human B-cell receptor specific CD4 + T cells. Oncoimmunology 2016; 5:e1232220. [PMID: 27999743 DOI: 10.1080/2162402x.2016.1232220] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 08/30/2016] [Accepted: 08/31/2016] [Indexed: 10/21/2022] Open
Abstract
The B-cell receptor (BCR) expressed by a clonal B cell tumor is a tumor specific antigen (idiotype). However, the T-cell epitopes within human BCRs which stimulate protective immunity still lack detailed characterization. In this study, we identified 17 BCR peptide-specific CD4+ T-cell epitopes derived from BCR heavy and light chain variable region sequences. Detailed analysis revealed these CD4+ T-cell epitopes stimulated normal donors' and patients' Th1 CD4+ T cells to directly recognize the autologous tumors by secretion of IFNγ, indicating the epitopes are processed and presented by tumor cells. One BCR peptide-specific CD4+ T cell line was also cytotoxic and lysed autologous tumor cells through the perforin pathway. Sequence analysis of the epitopes revealed that 10 were shared by multiple primary patients' tumors, and 16 had the capacity to bind to more than one HLA DRB1 allele. T cells stimulated by shared epitopes recognized primary tumors expressing the same sequences on multiple HLA DRB1 alleles. In conclusion, we identified 17 BCR-derived CD4+ T-cell epitopes with promiscuous HLA DRB1 binding affinity that are shared by up to 36% of patients, suggesting a strategy to overcome the requirement for individual preparation of therapeutic agents targeting idiotype.
Collapse
Affiliation(s)
- Jinsheng Weng
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Center Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Flavio Egidio Baio
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Kelsey E Moriarty
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Hiroki Torikai
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Hua Wang
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Zhiqiang Liu
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Sourindra N Maiti
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Dongho Gwak
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Michael S Popescu
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Soung-Chul Cha
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Laurence J N Cooper
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Sattva S Neelapu
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Larry W Kwak
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| |
Collapse
|
23
|
Oki S. Pathological mechanism of secondary-progressive multiples sclerosis and its animal model. ACTA ACUST UNITED AC 2016; 39:103-13. [PMID: 27212596 DOI: 10.2177/jsci.39.103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Development of acute experimental autoimmune encephalomyelitis (EAE) depends on Th17 cells expressing the nuclear factor NR4A2, which we have previously reported to be upregulated in peripheral blood T cells from patients of multiple sclerosis (MS). EAE induced in mice lacking NR4A2 in T cells showed a great reduction in Th17-mediated acute symptoms, whereas a late-onset disease independent of NR4A2 was still inducible. We identified cytotoxic T-cell-like CD4+ T cells expressing the T-box transcription factor Eomesodermin (Eomes) as a pathogenic component for the development of the late-onset disease. Furthermore, T cell-specific deletion of the Eomes gene or Eomes-specific RNA interference in vivo remarkably ameliorated the late-onset EAE. Intriguingly, similar Eomes-expressing CD4+ T cells are increased in the peripheral blood and cerebrospinal fluid only from patients with secondary-progressive MS accompanied by neurodegenerative symptoms, but not in relapsing-remitting MS. Mechanistic analysis revealed that granzyme B was secreted by Eomes-expressing CD4+ T cells and the activation of protease-activated receptor-1 by granzyme B is involved in the neuroinflammation observed in the late-onset EAE.
Collapse
Affiliation(s)
- Shinji Oki
- Department of Immunology, National Institute of Neuroscience, NCNP
| |
Collapse
|
24
|
Winchester R, Giles JT, Nativ S, Downer K, Zhang HZ, Bag-Ozbek A, Zartoshti A, Bokhari S, Bathon JM. Association of Elevations of Specific T Cell and Monocyte Subpopulations in Rheumatoid Arthritis With Subclinical Coronary Artery Atherosclerosis. Arthritis Rheumatol 2016; 68:92-102. [PMID: 26360530 DOI: 10.1002/art.39419] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Accepted: 08/27/2015] [Indexed: 01/20/2023]
Abstract
OBJECTIVE Coronary artery disease (CAD) is the leading cause of excess deaths in rheumatoid arthritis (RA). However, identification of features denoting patients with a risk of developing CAD is lacking. The composition of circulating peripheral blood mononuclear cell (PBMC) subsets in RA patients differs markedly from that in healthy controls with regard to the extent of T cell activation, with clonal expansion and differentiation to effector memory status, and presence of inflammatory monocytes. In this study, we sought to evaluate whether elevations in these PBMC subpopulations in RA patients could denote those with an increased risk of subclinical CAD, as determined by the presence of coronary artery calcification (CAC). METHODS The study cohort comprised 72 patients with RA who underwent cardiac computed tomography to assess CAC. PBMC subsets were determined by multiparameter flow cytometry. Multivariable logistic regression was used to determine the associations between PBMC subpopulations and the presence of CAC. RESULTS Among the 72 patients with RA, 33% had CAC and exhibited significant increases in the levels of circulating CD4 T cell subsets denoting activation and differentiation to the effector memory phenotypes. Analogous increases in the levels of CD8 T cell subsets, as well as in the CD14(high)CD16+ intermediate monocyte subset, were also present in these patients, as compared to those without CAC. The increases in the CD4 and CD8 T cell subsets were highly intercorrelated, whereas the increases in CD14(high)CD16+ monocytes were independent of elevations in the CD4 T cell subsets. After adjustments for relevant confounders, the levels of CD4+CD56+CD57+ T cells and CD14(high)CD16+ monocytes remained associated with the presence of CAC. CONCLUSION These findings indicate that PBMC subsets are markers for the presence of CAC and suggest that mechanisms of atherogenesis in RA may operate in part through the elevations in these subsets, raising further questions about the mechanisms underlying the presence of such alterations in cell composition in patients with RA and the potential for shared etiologic pathways between RA and cardiovascular disease.
Collapse
Affiliation(s)
- Robert Winchester
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Jon T Giles
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Simona Nativ
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Kendall Downer
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Hui-Zhu Zhang
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Ayse Bag-Ozbek
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Afshin Zartoshti
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Sabahat Bokhari
- Columbia University College of Physicians and Surgeons, New York, New York
| | - Joan M Bathon
- Columbia University College of Physicians and Surgeons, New York, New York
| |
Collapse
|
25
|
Brown DM, Lampe AT, Workman AM. The Differentiation and Protective Function of Cytolytic CD4 T Cells in Influenza Infection. Front Immunol 2016; 7:93. [PMID: 27014272 PMCID: PMC4783394 DOI: 10.3389/fimmu.2016.00093] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 02/25/2016] [Indexed: 12/24/2022] Open
Abstract
CD4 T cells that recognize peptide antigen in the context of class II MHC can differentiate into various subsets that are characterized by their helper functions. However, increasing evidence indicates that CD4 cells with direct cytolytic activity (CD4 CTL) play a role in chronic as well as acute infections, such as influenza A virus (IAV) infection. In the last couple of decades, techniques to measure the frequency and activity of these cytolytic cells has demonstrated their abundance in infections, such as human immunodeficiency virus, mouse pox, murine gamma herpes virus, cytomegalovirus, Epstein-Barr virus, and influenza among others. We now appreciate a greater role for CD4 CTL as direct effectors in viral infections and antitumor immunity through their ability to acquire perforin-mediated cytolytic activity and contribution to lysis of virally infected targets or tumors. As early as the 1980s, CD4 T cell clones with cytolytic potential were identified after influenza virus infection, yet much of this early work was dependent on in vitro culture and little was known about the physiological relevance of CD4 CTL. Here, we discuss the direct role CD4 CTL play in protection against lethal IAV infection and the factors that drive the generation of perforin-mediated lytic activity in CD4 cells in vivo during IAV infection. While focusing on CD4 CTL generated during IAV infection, we pull comparisons from the literature in other antiviral and antitumor systems. Further, we highlight what is currently known about CD4 CTL secondary and memory responses, as well as vaccination strategies to induce these potent killer cells that provide an extra layer of cell-mediated immune protection against heterosubtypic IAV infection.
Collapse
Affiliation(s)
- Deborah M Brown
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA; Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Anna T Lampe
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA; Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Aspen M Workman
- Nebraska Center for Virology, University of Nebraska-Lincoln , Lincoln, NE , USA
| |
Collapse
|
26
|
Wyluda EJ, Cheng J, Schell TD, Haley JS, Mallon C, Neves RI, Robertson G, Sivik J, Mackley H, Talamo G, Drabick JJ. Durable complete responses off all treatment in patients with metastatic malignant melanoma after sequential immunotherapy followed by a finite course of BRAF inhibitor therapy. Cancer Biol Ther 2016; 16:662-70. [PMID: 25806780 PMCID: PMC4622667 DOI: 10.1080/15384047.2015.1026507] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
We report 3 cases of durable complete response (CR) in patients with BRAF-mutated metastatic melanoma who were initially treated unsuccessfully with sequential immunotherapies (high dose interleukin 2 followed by ipilimumab with or without concurrent radiation therapy). After progression during or post immunotherapy, these patients were given BRAF inhibitor therapy and developed rapid CRs. Based on the concomitant presence of autoimmune manifestations (including vitiligo and hypophysitis), we postulated that there was a synergistic effect between the prior immune therapy and the BRAF targeting agents. Accordingly, the inhibitors were gradually weaned off beginning at 3 months and were stopped completely at 9-12 months. The three patients remain well and in CR off of all therapy at up to 15 months radiographic follow-up. The institution of the BRAF therapy was associated with development of severe rheumatoid-like arthritis in 2 patients which persisted for months after discontinuation of therapy, suggesting it was not merely a known toxicity of BRAF inhibitors (arthralgias). On immunologic analysis, these patients had high levels of non-T-regulatory, CD4 positive effector phenotype T-cells, which persisted after completion of therapy. Of note, we had previously reported a similar phenomenon in patients with metastatic melanoma who failed high dose interleukin-2 and were then placed on a finite course of temozolomide with rapid complete responses that have remained durable for many years after discontinuation of temozolomide. We postulate that a finite course of cytotoxic or targeted therapy specific for melanoma given after apparent failure of prior immunotherapy can result in complete and durable remissions that may persist long after the specific cytotoxic or targeted agents have been discontinued suggesting the existence of sequence specific synergism between immunotherapy and these agents. Here, we discuss these cases in the context of the literature on synergy between conventional or targeted cytotoxic therapy and immunotherapy in cancer treatment.
Collapse
Key Words
- BRAF inhibitor
- CBC, complete blood count
- CR, complete response
- CRP, c-reactive protein
- CT, computed tomography
- CTL, cytotoxic lymphocyte
- CTLA-4, cytotoxic T-lymphocyte-associated protein 4
- GrzB, granzyme B
- HD, high dose
- IFN, interferon
- IL-2, interleukin 2
- LDH, lactate dehydrogenase
- M6P, manose 6 phosphate
- MAPK, mitogen-activated protein kinase pathway
- PD-1, programmed death 1
- PDL-1, programmed death ligand 1
- PDL-2, programmed death ligand 2
- PET, positron emission tomography
- PR, partial response
- RT, radiation therapy
- SLE, systemic lupus erythematosus
- WBC, white blood cell count
- cytotoxic therapy, immunotherapy, treatment of melanoma
- interleukin-2
- ipilimumab
- metastatic melanoma
Collapse
Affiliation(s)
- Edward J Wyluda
- a Division of Hematology Oncology; Penn State Milton S Hershey Medical Center ; Hershey , PA , USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Taylor EB, Moulana M, Stuge TB, Quiniou SMA, Bengten E, Wilson M. A Leukocyte Immune-Type Receptor Subset Is a Marker of Antiviral Cytotoxic Cells in Channel Catfish, Ictalurus punctatus. THE JOURNAL OF IMMUNOLOGY 2016; 196:2677-89. [PMID: 26856701 DOI: 10.4049/jimmunol.1502166] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 01/03/2016] [Indexed: 11/19/2022]
Abstract
Channel catfish, Ictalurus punctatus, leukocyte immune type receptors (LITRs) represent a multigene family that encodes Ig superfamily proteins that mediate activating or inhibitory signaling. In this study, we demonstrate the use of mAb CC41 to monitor viral cytotoxic responses in catfish and determine that CC41 binds to a subset of LITRs on the surface of catfish clonal CTLs. Homozygous gynogenetic catfish were immunized with channel catfish virus (CCV)-infected MHC-matched clonal T cells (G14D-CCV), and PBL were collected at various times after immunization for flow cytometric analyses. The percentage of CC41(+) cells was significantly increased 5 d after primary immunization with G14D-CCV and at 3 d after a booster immunization as compared with control fish only injected with G14D. Moreover, CC41(+) cells magnetically isolated from the PBL specifically killed CCV-infected targets as measured by (51)Cr release assays and expressed messages for CD3γδ, perforin, and at least one of the CD4-like receptors as analyzed by RNA flow cytometry. When MLC effector cells derived from a G14D-CCV-immunized fish were preincubated with CC41 mAb, killing of G14D-CCV targets was reduced by ∼40%, suggesting that at least some LITRs have a role in target cell recognition and/or cytotoxicity. The availability of a LITR-specific mAb has allowed, to our knowledge for the first time, functional characterization of LITRs in an autologous system. In addition, the identification of an LITR subset as a cytotoxic cell marker will allow for more effective monitoring of catfish immune responses to pathogens.
Collapse
Affiliation(s)
- Erin B Taylor
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Mohadetheh Moulana
- Warmwater Aquaculture Research Unit, U.S. Department of Agriculture-Agricultural Research Service, Stoneville, MS 38776; and
| | - Tor B Stuge
- Immunology Research Group, Department of Medical Biology, Faculty of Health Sciences, University of Tromso-Arctic University of Norway, N-9037 Tromso, Norway
| | - Sylvie M A Quiniou
- Warmwater Aquaculture Research Unit, U.S. Department of Agriculture-Agricultural Research Service, Stoneville, MS 38776; and
| | - Eva Bengten
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, MS 39216
| | - Melanie Wilson
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, MS 39216;
| |
Collapse
|
28
|
Riaz T, Sollid LM, Olsen I, de Souza GA. Quantitative Proteomics of Gut-Derived Th1 and Th1/Th17 Clones Reveal the Presence of CD28+ NKG2D- Th1 Cytotoxic CD4+ T cells. Mol Cell Proteomics 2015; 15:1007-16. [PMID: 26637539 DOI: 10.1074/mcp.m115.050138] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Indexed: 12/13/2022] Open
Abstract
T-helper cells are differentiated from CD4+ T cells and are traditionally characterized by inflammatory or immunosuppressive responses in contrast to cytotoxic CD8+ T cells. Mass-spectrometry studies on T-helper cells are rare. In this study, we aimed to identify the proteomes of human Th1 and Th1/Th17 clones derived from intestinal biopsies of Crohn's disease patients and to identify differentially expressed proteins between the two phenotypes. Crohn's disease is an inflammatory bowel disease, with predominantly Th1- and Th17-mediated response where cells of the "mixed" phenotype Th1/Th17 have also been commonly found. High-resolution mass spectrometry was used for protein identification and quantitation. In total, we identified 7401 proteins from Th1 and Th1/Th17 clones, where 334 proteins were differentially expressed. Major differences were observed in cytotoxic proteins that were overrepresented in the Th1 clones. The findings were validated by flow cytometry analyses using staining with anti-granzyme B and anti-perforin and by a degranulation assay, confirming higher cytotoxic features of Th1 compared with Th1/Th17 clones. By testing a larger panel of T-helper cell clones from seven different Crohn's disease patients, we concluded that only a subgroup of the Th1 cell clones had cytotoxic features, and these expressed the surface markers T-cell-specific surface glycoprotein CD28 and were negative for expression of natural killer group 2 member D.
Collapse
Affiliation(s)
- Tahira Riaz
- From the Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, 0372 Oslo, Norway;
| | - Ludvig Magne Sollid
- From the Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, 0372 Oslo, Norway
| | - Ingrid Olsen
- From the Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, 0372 Oslo, Norway; Section for Immunology, Norwegian Veterinary Institute, Ullevaalsveien 68, 0454 Oslo, Norway
| | - Gustavo Antonio de Souza
- From the Centre for Immune Regulation and Department of Immunology, University of Oslo and Oslo University Hospital-Rikshospitalet, 0372 Oslo, Norway
| |
Collapse
|
29
|
Bradfute SB, Anthony SM, Stuthman KS, Ayithan N, Tailor P, Shaia CI, Bray M, Ozato K, Bavari S. Mechanisms of immunity in post-exposure vaccination against Ebola virus infection. PLoS One 2015; 10:e0118434. [PMID: 25785602 PMCID: PMC4364937 DOI: 10.1371/journal.pone.0118434] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 01/16/2015] [Indexed: 01/07/2023] Open
Abstract
Ebolaviruses can cause severe hemorrhagic fever that is characterized by rapid viral replication, coagulopathy, inflammation, and high lethality rates. Although there is no clinically proven vaccine or treatment for Ebola virus infection, a virus-like particle (VLP) vaccine is effective in mice, guinea pigs, and non-human primates when given pre-infection. In this work, we report that VLPs protect Ebola virus-infected mice when given 24 hours post-infection. Analysis of cytokine expression in serum revealed a decrease in pro-inflammatory cytokine and chemokine levels in mice given VLPs post-exposure compared to infected, untreated mice. Using knockout mice, we show that VLP-mediated post-exposure protection requires perforin, B cells, macrophages, conventional dendritic cells (cDCs), and either CD4+ or CD8+ T cells. Protection was Ebola virus-specific, as marburgvirus VLPs did not protect Ebola virus-infected mice. Increased antibody production in VLP-treated mice correlated with protection, and macrophages were required for this increased production. However, NK cells, IFN-gamma, and TNF-alpha were not required for post-exposure-mediated protection. These data suggest that a non-replicating Ebola virus vaccine can provide post-exposure protection and that the mechanisms of immune protection in this setting require both increased antibody production and generation of cytotoxic T cells.
Collapse
Affiliation(s)
- Steven B. Bradfute
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Scott M. Anthony
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Kelly S. Stuthman
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Natarajan Ayithan
- Laboratory of Molecular Growth Regulation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | | | - Carl I. Shaia
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Mike Bray
- Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Keiko Ozato
- Laboratory of Molecular Growth Regulation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Sina Bavari
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
- * E-mail:
| |
Collapse
|
30
|
Lintermans LL, Stegeman CA, Heeringa P, Abdulahad WH. T cells in vascular inflammatory diseases. Front Immunol 2014; 5:504. [PMID: 25352848 PMCID: PMC4196542 DOI: 10.3389/fimmu.2014.00504] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 09/28/2014] [Indexed: 12/12/2022] Open
Abstract
Inflammation of the human vasculature is a manifestation of many different diseases ranging from systemic autoimmune diseases to chronic inflammatory diseases, in which multiple types of immune cells are involved. For both autoimmune diseases and chronic inflammatory diseases several observations support a key role for T lymphocytes in these disease pathologies, but the underlying mechanisms are poorly understood. Previous studies in several autoimmune diseases have demonstrated a significant role for a specific subset of CD4+ T cells termed effector memory T (TEM) cells. This expanded population of TEM cells may contribute to tissue injury and disease progression. These cells exert multiple pro-inflammatory functions through the release of effector cytokines. Many of these cytokines have been detected in the inflammatory lesions and participate in the vasculitic reaction, contributing to recruitment of macrophages, neutrophils, dendritic cells, natural killer cells, B cells, and T cells. In addition, functional impairment of regulatory T cells paralyzes anti-inflammatory effects in vasculitic disorders. Interestingly, activation of TEM cells is uniquely dependent on the voltage-gated potassium Kv1.3 channel providing an anchor for specific drug targeting. In this review, we focus on the CD4+ T cells in the context of vascular inflammation and describe the evidence supporting the role of different T cell subsets in vascular inflammation. Selective targeting of pathogenic TEM cells might enable a more tailored therapeutic approach that avoids unwanted adverse side effects of generalized immunosuppression by modulating the effector functions of T cell responses to inhibit the development of vascular inflammation.
Collapse
Affiliation(s)
- Lucas L Lintermans
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen , Groningen , Netherlands
| | - Coen A Stegeman
- Department of Nephrology, University of Groningen, University Medical Center Groningen , Groningen , Netherlands
| | - Peter Heeringa
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen , Groningen , Netherlands
| | - Wayel H Abdulahad
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen , Groningen , Netherlands
| |
Collapse
|
31
|
Siciliano NA, Hersperger AR, Lacuanan AM, Xu RH, Sidney J, Sette A, Sigal LJ, Eisenlohr LC. Impact of distinct poxvirus infections on the specificities and functionalities of CD4+ T cell responses. J Virol 2014; 88:10078-91. [PMID: 24965457 PMCID: PMC4136331 DOI: 10.1128/jvi.01150-14] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2014] [Accepted: 06/13/2014] [Indexed: 12/14/2022] Open
Abstract
UNLABELLED The factors that determine CD4+ T cell (TCD4+) specificities, functional capacity, and memory persistence in response to complex pathogens remain unclear. We explored these parameters in the C57BL/6 mouse through comparison of two highly related (>92% homology) poxviruses: ectromelia virus (ECTV), a natural mouse pathogen, and vaccinia virus (VACV), a heterologous virus that nevertheless elicits potent immune responses. In addition to elucidating several previously unidentified major histocompatibility complex class II (MHC-II)-restricted epitopes, we observed many qualitative and quantitative differences between the TCD4+ repertoires, including responses not elicited by VACV despite complete sequence conservation. In addition, we observed functional heterogeneity between ECTV- and VACV-specific TCD4+ at both a global and individual epitope level, particularly greater expression of the cytolytic marker CD107a from TCD4+ following ECTV infection. Most striking were differences during the late memory phase where, in contrast to ECTV, VACV infection failed to elicit measurable epitope-specific TCD4+ as determined by intracellular cytokine staining. These findings illustrate the strong influence of epitope-extrinsic factors on TCD4+ responses and memory. IMPORTANCE Much of our understanding concerning host-pathogen relationships in the context of poxvirus infections stems from studies of VACV in mice. However, VACV is not a natural mouse pathogen, and therefore, the relevance of results obtained using this model may be limited. Here, we explored the MHC class II-restricted TCD4+ repertoire induced by mousepox (ECTV) infection and the functional profile of the responding epitope-specific TCD4+, comparing these results to those induced by VACV infection under matched conditions. Despite a high degree of homology between the two viruses, we observed distinct specificity and functional profiles of TCD4+ responses at both acute and memory time points, with VACV-specific TCD4+ memory being notably compromised. These data offer insight into the impact of epitope-extrinsic factors on the resulting TCD4+ responses.
Collapse
Affiliation(s)
- Nicholas A Siciliano
- Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Adam R Hersperger
- Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA Department of Biology, Albright College, Reading, Pennsylvania, USA
| | - Aimee M Lacuanan
- Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Ren-Huan Xu
- Fox Chase Cancer Center, Immune Cell Development and Host Defense Program, Philadelphia, Pennsylvania, USA
| | - John Sidney
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Luis J Sigal
- Fox Chase Cancer Center, Immune Cell Development and Host Defense Program, Philadelphia, Pennsylvania, USA
| | - Laurence C Eisenlohr
- Department of Microbiology and Immunology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
32
|
Croia C, Astorri E, Murray-Brown W, Willis A, Brokstad KA, Sutcliffe N, Piper K, Jonsson R, Tappuni AR, Pitzalis C, Bombardieri M. Implication of Epstein-Barr Virus Infection in Disease-Specific Autoreactive B Cell Activation in Ectopic Lymphoid Structures of Sjögren's Syndrome. Arthritis Rheumatol 2014; 66:2545-57. [DOI: 10.1002/art.38726] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Accepted: 05/22/2014] [Indexed: 12/11/2022]
Affiliation(s)
- Cristina Croia
- William Harvey Research Institute, Queen Mary University of London; London UK
| | - Elisa Astorri
- William Harvey Research Institute, Queen Mary University of London; London UK
| | | | - Amanda Willis
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London; London UK
| | | | - Nurhan Sutcliffe
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London; London UK
| | - Kim Piper
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London; London UK
| | | | - Anwar R. Tappuni
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London; London UK
| | - Costantino Pitzalis
- William Harvey Research Institute, Queen Mary University of London; London UK
| | - Michele Bombardieri
- William Harvey Research Institute, Queen Mary University of London; London UK
| |
Collapse
|
33
|
Saez-Cirion A, Jacquelin B, Barré-Sinoussi F, Müller-Trutwin M. Immune responses during spontaneous control of HIV and AIDS: what is the hope for a cure? Philos Trans R Soc Lond B Biol Sci 2014; 369:20130436. [PMID: 24821922 PMCID: PMC4024229 DOI: 10.1098/rstb.2013.0436] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
HIV research has made rapid progress and led to remarkable achievements in recent decades, the most important of which are combination antiretroviral therapies (cART). However, in the absence of a vaccine, the pandemic continues, and additional strategies are needed. The 'towards an HIV cure' initiative aims to eradicate HIV or at least bring about a lasting remission of infection during which the host can control viral replication in the absence of cART. Cases of spontaneous and treatment-induced control of infection offer substantial hope. Here, we describe the scientific knowledge that is lacking, and the priorities that have been established for research into a cure. We discuss in detail the immunological lessons that can be learned by studying natural human and animal models of protection and spontaneous control of viraemia or of disease progression. In particular, we describe the insights we have gained into the immune mechanisms of virus control, the impact of early virus-host interactions and why chronic inflammation, a hallmark of HIV infection, is an obstacle to a cure. Finally, we enumerate current interventions aimed towards improving the host immune response.
Collapse
Affiliation(s)
| | | | | | - M. Müller-Trutwin
- Institut Pasteur, Unité de Régulation des Infections Rétrovirales, Paris, France
| |
Collapse
|
34
|
Abstract
OBJECTIVES Immune factors determining clinical progression following HIV-1 infection remain unclear. The SPARTAC trial randomized 366 participants in primary HIV infection (PHI) to different short-course therapies. The aim of this study was to investigate how early immune responses in PHI impacted clinical progression in SPARTAC. DESIGN AND METHODS Participants with PHI recruited to the SPARTAC trial were sampled at enrolment, prior to commencing any therapy. HIV-1-specific CD4(+) and CD8(+) ELISpot responses were measured by gamma interferon ELISPOT. Immunological data were associated with baseline covariates and times to clinical progression using logistic regression, Kaplan-Meier plots, and Cox models. RESULTS Making a CD4(+) T-cell ELISpot response (n = 119) at enrolment was associated with higher CD4(+) cell counts (P = 0.02) and to some extent lower plasma HIV RNA (P = 0.07). There was no correlation between the number of overlapping Gag CD8(+) T-cell ELISpot responses (n = 138) and plasma HIV-1 RNA viral load. Over a median follow-up of 2.9 years, baseline CD4(+) cell ELISpot responses (n = 119) were associated with slower clinical progression (P = 0.01; log-rank). Over a median of 3.1 years, there was no evidence for a survival advantage imposed by CD8(+) T-cell immunity (P = 0.82). CONCLUSION These data support a dominant protective role for CD4(+) T-cell immunity in PHI compared with CD8(+) T-cell responses, and are highly pertinent to HIV pathogenesis and vaccines, indicating that vaccine-induced CD4(+) responses may confer sustained benefit.
Collapse
|
35
|
Workman AM, Jacobs AK, Vogel AJ, Condon S, Brown DM. Inflammation enhances IL-2 driven differentiation of cytolytic CD4 T cells. PLoS One 2014; 9:e89010. [PMID: 24586481 PMCID: PMC3930678 DOI: 10.1371/journal.pone.0089010] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Accepted: 01/14/2014] [Indexed: 12/24/2022] Open
Abstract
Cytolytic CD4 T cells (CD4 CTL) have been identified in vivo in response to viral infections; however, the factors necessary for driving the cytolytic phenotype have not been fully elucidated. Our previously published work suggests IL-2 may be the master regulator of perforin-mediated cytotoxicity in CD4 effectors. To further dissect the role of IL-2 in CD4 CTL generation, T cell receptor transgenic mice deficient in the ability to produce IL-2 or the high affinity IL-2 receptor (IL-2Rα, CD25) were used. Increasing concentrations of IL-2 were necessary to drive perforin (Prf) expression and maximal cytotoxicity. Granzyme B (GrB) expression and killing correlated with STAT5 activation and CD25 expression in vitro, suggesting that signaling through the high affinity IL-2R is critical for full cytotoxicity. IL-2 signaling was also necessary in vivo for inducing the Th1 phenotype and IFN-γ expression in CD4 T cells during influenza A (IAV) infection. In addition, GrB expression, as measured by mean fluorescent intensity, was decreased in CD25 deficient cells; however, the frequency of CD4 cells expressing GrB was unchanged. Similarly, analysis of cytolytic markers such as CD107a/b and Eomesodermin indicate high IL-2Rα expression is not necessary to drive the CD4 CTL phenotype during IAV infection. Thus, inflammatory signals induced by viral infection may overcome the need for strong IL-2 signals in driving cytotoxicity in CD4 cells.
Collapse
Affiliation(s)
- Aspen M Workman
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America ; Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Ashley K Jacobs
- Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Alexander J Vogel
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Shirley Condon
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America ; Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Deborah M Brown
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America ; Nebraska Center for Virology, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| |
Collapse
|
36
|
Rambal V, Müller K, Dang-Heine C, Sattler A, Dziubianau M, Weist B, Luu SH, Stoyanova A, Nickel P, Thiel A, Neumann A, Schweiger B, Reinke P, Babel N. Differential influenza H1N1-specific humoral and cellular response kinetics in kidney transplant patients. Med Microbiol Immunol 2014; 203:35-45. [PMID: 24057515 DOI: 10.1007/s00430-013-0312-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 09/03/2013] [Indexed: 12/17/2022]
Abstract
Renal transplant recipients (RTR) are considered at high risk for influenza-associated complications due to immunosuppression. The efficacy of standard influenza vaccination in RTRs is unclear. Hence, we evaluated activation of the adaptive immunity by the pandemic influenza A(H1N1) 2009 (A(H1N1)pdm09) vaccine in RTRs as compared to healthy controls. To determine cross-reactivity and/or bystander activation, seasonal trivalent influenza vaccine and tetanus/diphteria toxoid (TT/DT) vaccine-specific T cells along with allospecific T cells were quantified before and after A(H1N1)pdm09 vaccination. Vaccination-induced alloimmunity was additionally determined by quantifying serum creatinine and proinflammatory protein IP-10. Contrary to healthy controls, RTRs required a booster vaccination to achieve seroconversion (13.3 % day 21; 90 % day 90). In contrast to humoral immunity, sufficient A(H1N1)pdm09-specific T-cell responses were mounted in RTRs already after the first immunization with a magnitude comparable with healthy controls. Interestingly, vaccination simultaneously boosted T cells reacting to seasonal flu but not to TT/DT, suggesting cross-activation. No alloimmune effects were recorded. In conclusion, protective antibody responses required booster vaccination. However, sufficient cellular immunity is established already after the first vaccination, demonstrating differential kinetics of humoral and cellular immunity.
Collapse
Affiliation(s)
- Vinay Rambal
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité University Medicine Berlin, Campus Virchow Clinic, Augustenburger Platz 1, 13353, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
|
38
|
Martirosyan A, Von Bargen K, Arce Gorvel V, Zhao W, Hanniffy S, Bonnardel J, Méresse S, Gorvel JP. In vivo identification and characterization of CD4⁺ cytotoxic T cells induced by virulent Brucella abortus infection. PLoS One 2013; 8:e82508. [PMID: 24367519 PMCID: PMC3868576 DOI: 10.1371/journal.pone.0082508] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 10/24/2013] [Indexed: 12/20/2022] Open
Abstract
CD4+ T cells display a variety of helper functions necessary for an efficient adaptive immune response against bacterial invaders. This work reports the in vivo identification and characterization of murine cytotoxic CD4+ T cells (CD4+ CTL) during Brucella abortus infection. These CD4+ CTLs express granzyme B and exhibit immunophenotypic features consistent with fully differentiated T cells. They express CD25, CD44, CD62L ,CD43 molecules at their surface and produce IFN-γ. Moreover, these cells express neither the co-stimulatory molecule CD27 nor the memory T cell marker CD127. We show here that CD4+ CTLs are capable of cytolytic action against Brucella-infected antigen presenting cells (APC) but not against Mycobacterium-infected APC. Cytotoxic CD4+ T cell population appears at early stages of the infection concomitantly with high levels of IFN-γ and granzyme B expression. CD4+ CTLs represent a so far uncharacterized immune cell sub-type triggered by early immune responses upon Brucella abortus infection.
Collapse
Affiliation(s)
- Anna Martirosyan
- Aix-Marseille University, CIML, 13288, Marseille, France
- CNRS, UMR 7280, 13288, Marseille, France
- INSERM, U631, 13288, Marseille, France
| | - Kristine Von Bargen
- Aix-Marseille University, CIML, 13288, Marseille, France
- CNRS, UMR 7280, 13288, Marseille, France
- INSERM, U631, 13288, Marseille, France
| | - Vilma Arce Gorvel
- Aix-Marseille University, CIML, 13288, Marseille, France
- CNRS, UMR 7280, 13288, Marseille, France
- INSERM, U631, 13288, Marseille, France
| | - Weidong Zhao
- Aix-Marseille University, CIML, 13288, Marseille, France
- CNRS, UMR 7280, 13288, Marseille, France
- INSERM, U631, 13288, Marseille, France
| | - Sean Hanniffy
- Aix-Marseille University, CIML, 13288, Marseille, France
- CNRS, UMR 7280, 13288, Marseille, France
- INSERM, U631, 13288, Marseille, France
| | - Johnny Bonnardel
- Aix-Marseille University, CIML, 13288, Marseille, France
- CNRS, UMR 7280, 13288, Marseille, France
- INSERM, U631, 13288, Marseille, France
| | - Stéphane Méresse
- Aix-Marseille University, CIML, 13288, Marseille, France
- CNRS, UMR 7280, 13288, Marseille, France
- INSERM, U631, 13288, Marseille, France
| | - Jean-Pierre Gorvel
- Aix-Marseille University, CIML, 13288, Marseille, France
- CNRS, UMR 7280, 13288, Marseille, France
- INSERM, U631, 13288, Marseille, France
- *
| |
Collapse
|
39
|
Tumor protective activity of CD4+ but not of CD8+ T cells in DNA-vaccinated mice challenged with bcr-abl-transformed cells. Clin Dev Immunol 2013; 2013:923107. [PMID: 24348684 PMCID: PMC3856117 DOI: 10.1155/2013/923107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 10/31/2013] [Accepted: 11/02/2013] [Indexed: 11/18/2022]
Abstract
In the recent past, it has repeatedly been reported that CD4 cells play an important role in the immunology of chronic myeloid leukaemia. It was therefore of interest to test their activity in an animal model using bcr-abl-transformed cells. BALB/c mice were four times immunized with a DNA vaccine carrying the bcr-abl fusion gene. Two weeks after the last vaccine dose, the animals were challenged with syngeneic bcr-abl-transformed 12B1 cells which form solid tumors after subcutaneous administration. At the time of challenge, animals were treated with antibodies against the CD8+ T cells or CD4+ T cells. The efficacy of the depletion was monitored and found highly effective. All nonimmunized animals developed tumors. All animals untreated with the antibodies as well as those in which CD8+ T cells had been depleted, were fully protected against the challenge. On the other hand, almost all mice treated with anti-CD4+ antibody developed tumors. These results strongly suggested that the CD4+ T cells acted as effectors in the present system.
Collapse
|
40
|
|
41
|
Croia C, Serafini B, Bombardieri M, Kelly S, Humby F, Severa M, Rizzo F, Coccia EM, Migliorini P, Aloisi F, Pitzalis C. Epstein-Barr virus persistence and infection of autoreactive plasma cells in synovial lymphoid structures in rheumatoid arthritis. Ann Rheum Dis 2013; 72:1559-68. [PMID: 23268369 DOI: 10.1136/annrheumdis-2012-202352] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Rheumatoid arthritis (RA) is associated with an increased Epstein-Barr virus (EBV) blood DNA load, a robust immune response to EBV and cross-reactive circulating antibodies to viral and self-antigens. However, the role of EBV in RA pathogenesis remains elusive. Here, we investigated the relationship between synovial EBV infection, ectopic lymphoid structures (ELS) and immunity to citrullinated self and EBV proteins. METHODS Latent and lytic EBV infection was investigated in 43 RA synovial tissues characterised for presence/absence of ELS and in 11 control osteoarthritis synovia using RT-PCR, in situ hybridisation and immunohistochemistry. Synovial production of anti-citrullinated protein (ACPA) and anti-citrullinated EBV peptide (VCP1/VCP2) antibodies was investigated in situ and in vivo in the severe combined immunodeficiency (SCID)/RA chimeric model. RESULTS EBV dysregulation was observed exclusively in ELS+ RA but not osteoarthritis (OA) synovia, as revealed by presence of EBV latent (LMP2A, EBV-encoded small RNA (EBER)) transcripts, EBER+ cells and immunoreactivity for EBV latent (LMP1, LMP2A) and lytic (BFRF1) antigens in ELS-associated B cells and plasma cells, respectively. Importantly, a large proportion of ACPA-producing plasma cells surrounding synovial germinal centres were infected with EBV. Furthermore, ELS-containing RA synovia transplanted into SCID mice supported production of ACPA and anti-VCP1/VCP2 antibodies. Analysis of CD4+ and CD8+ T-cell localisation and granzyme B expression suggests that EBV persistence in ELS-containing synovia may be favoured by exclusion of CD8+ T cells from B-cell follicles and impaired CD8-mediated cytotoxicity. CONCLUSIONS We demonstrated active EBV infection within ELS in the RA synovium in association with local differentiation of ACPA-reactive B cells.
Collapse
Affiliation(s)
- Cristina Croia
- Centre for Experimental Medicine and Rheumatology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Cytokine-dependent induction of CD4+ T cells with cytotoxic potential during influenza virus infection. J Virol 2013; 87:11884-93. [PMID: 23986597 DOI: 10.1128/jvi.01461-13] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Recent evidence has identified the role of granzyme B- and perforin-expressing CD4(+) T cells with cytotoxic potential in antiviral immunity. However, the in vivo cytokine cues and downstream pathways governing the differentiation of these cells are unclear. Here, we have identified that CD4(+) T cells with cytotoxic potential are specifically induced at the site of infection during influenza virus infection. The development of CD4(+) T cells with cytotoxic potential in vivo was dependent on the cooperation of the STAT2-dependent type I interferon signaling and the interleukin-2/interleukin-2 receptor alpha pathway for the induction of the transcription factors T-bet and Blimp-1. We showed that Blimp-1 promoted the binding of T-bet to the promoters of cytolytic genes in CD4(+) T cells and was required for the cytolytic function of the in vitro- and in vivo-generated CD4(+) T cells with cytotoxic potential. Thus, our data define the molecular basis of regulation of the in vivo development of this functionally cytotoxic Th subset during acute respiratory virus infection. The potential implications for the functions of these cells are discussed.
Collapse
|
43
|
Fu J, Zhang Z, Zhou L, Qi Z, Xing S, Lv J, Shi J, Fu B, Liu Z, Zhang JY, Jin L, Zhao Y, Lau GKK, Zhao J, Wang FS. Impairment of CD4+ cytotoxic T cells predicts poor survival and high recurrence rates in patients with hepatocellular carcinoma. Hepatology 2013; 58:139-49. [PMID: 22961630 DOI: 10.1002/hep.26054] [Citation(s) in RCA: 148] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2011] [Accepted: 08/24/2012] [Indexed: 01/10/2023]
Abstract
UNLABELLED The role of CD4(+) cytotoxic T cells (CTLs) in hepatocellular carcinoma (HCC) remains obscure. This study characterized CD4(+) CTLs in HCC patients and further elucidated the associations between CD4(+) CTLs and HCC disease progression. In all, 547 HCC patients, 44 chronic hepatitis B (CHB) patients, 86 liver cirrhosis (LC) patients, and 88 healthy individuals were enrolled in the study. CD4(+) CTLs were defined by flow cytometry, immunohistochemistry, and lytic granule exocytosis assays. A multivariate analysis of prognostic factors for overall survival was performed using the Cox proportional hazards model. Circulating and liver-infiltrating CD4(+) CTLs were found to be significantly increased in HCC patients during early stage disease, but decreased in progressive stages of HCC. This loss of CD4(+) CTLs was significantly correlated with high mortality rates and reduced survival time of HCC patients. In addition, the proliferation, degranulation, and production of granzyme A, granzyme B, and perforin of CD4(+) CTLs were inhibited by the increased forkhead/winged helix transcription factor (FoxP3(+) ) regulatory T cells in these HCC patients. Further analysis showed that both circulating and tumor-infiltrating CD4(+) CTLs were independent predictors of disease-free survival and overall survival after the resection of the HCC. CONCLUSION The progressive deficit in CD4(+) CTLs induced by increased FoxP3(+) regulatory T cells was correlated with poor survival and high recurrence rates in HCC patients. These data suggest that CD4(+) CTLs may represent both a potential prognostic marker and a therapeutic target for the treatment of HCC.
Collapse
Affiliation(s)
- Junliang Fu
- Research Center for Biological Therapy, Beijing 302 Hospital, Peking University Health Science Center, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Hildemann SK, Eberlein J, Davenport B, Nguyen TT, Victorino F, Homann D. High efficiency of antiviral CD4(+) killer T cells. PLoS One 2013; 8:e60420. [PMID: 23565245 PMCID: PMC3614903 DOI: 10.1371/journal.pone.0060420] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Accepted: 01/10/2013] [Indexed: 12/22/2022] Open
Abstract
The destruction of infected cells by cytotxic T lymphocytes (CTL) is integral to the effective control of viral and bacterial diseases, and CTL function at large has long been regarded as a distinctive property of the CD8(+)T cell subset. In contrast, and despite their first description more than three decades ago, the precise contribution of cytotoxic CD4(+)T cells to the resolution of infectious diseases has remained a matter of debate. In particular, the CTL activity of pathogen-specific CD4(+) "helper" T cells constitutes a single trait among a diverse array of other T cell functionalities, and overall appears considerably weaker than the cytolytic capacity of CD8(+) effector T cells. Here, using an in vivo CTL assay, we report that cytotoxic CD4(+)T cells are readily generated against both viral and bacterial pathogens, and that the efficiency of MHC-II-restricted CD4(+)T cell killing adjusted for effector:target cell ratios, precise specificities and functional avidities is comparable in magnitude to that of CD8(+)T cells. In fact, the only difference between specific CD4(+) and CD8(+)T cells pertains to the slightly delayed killing kinetics of the former demonstrating that potent CTL function is a cardinal property of both antiviral CD8(+) and CD4(+)T cells.
Collapse
Affiliation(s)
- Steven K. Hildemann
- University Clinic for Cardiology and Angiology I, University Heart Center, Freiburg-Bad Krozingen, Germany
- Merck Research Laboratories/MSD Global Clinical Trial Operations, Haar, Germany
| | - Jens Eberlein
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Bennett Davenport
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America
- Integrated Department of Immunology, University of Colorado Denver and National Jewish Health, Denver, Colorado, United States of America
- Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Tom T. Nguyen
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America
- Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Francisco Victorino
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America
- Integrated Department of Immunology, University of Colorado Denver and National Jewish Health, Denver, Colorado, United States of America
| | - Dirk Homann
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America
- Integrated Department of Immunology, University of Colorado Denver and National Jewish Health, Denver, Colorado, United States of America
- Department of Anesthesiology, University of Colorado Denver, Aurora, Colorado, United States of America
| |
Collapse
|
45
|
Abdulahad WH, De Souza AWS, Kallenberg CGM. L3. Are mononuclear cells predominant actors of endothelial damage in vasculitis? Presse Med 2013; 42:499-503. [PMID: 23477715 DOI: 10.1016/j.lpm.2013.02.305] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Affiliation(s)
- Wayel H Abdulahad
- University of Groningen, University Medical Center Groningen, Department of Rheumatology, Groningen, Netherlands.
| | | | | |
Collapse
|
46
|
Mucida D, Husain MM, Muroi S, van Wijk F, Shinnakasu R, Naoe Y, Reis BS, Huang Y, Lambolez F, Docherty M, Attinger A, Shui JW, Kim G, Lena CJ, Sakaguchi S, Miyamoto C, Wang P, Atarashi K, Park Y, Nakayama T, Honda K, Ellmeier W, Kronenberg M, Taniuchi I, Cheroutre H. Transcriptional reprogramming of mature CD4⁺ helper T cells generates distinct MHC class II-restricted cytotoxic T lymphocytes. Nat Immunol 2013; 14:281-9. [PMID: 23334788 PMCID: PMC3581083 DOI: 10.1038/ni.2523] [Citation(s) in RCA: 278] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2012] [Accepted: 12/14/2012] [Indexed: 12/12/2022]
Abstract
TCRαβ thymocytes differentiate into either CD8αβ(+) cytotoxic T lymphocytes or CD4(+) helper T cells. This functional dichotomy is controlled by key transcription factors, including the helper T cell master regulator ThPOK, which suppresses the cytolytic program in major histocompatibility complex (MHC) class II-restricted CD4(+) thymocytes. ThPOK continues to repress genes of the CD8 lineage in mature CD4(+) T cells, even as they differentiate into effector helper T cell subsets. Here we found that the helper T cell fate was not fixed and that mature, antigen-stimulated CD4(+) T cells terminated expression of the gene encoding ThPOK and reactivated genes of the CD8 lineage. This unexpected plasticity resulted in the post-thymic termination of the helper T cell program and the functional differentiation of distinct MHC class II-restricted CD4(+) cytotoxic T lymphocytes.
Collapse
Affiliation(s)
- Daniel Mucida
- Division of Developmental Immunology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| | - Mohammad Mushtaq Husain
- Division of Developmental Immunology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| | - Sawako Muroi
- Laboratory for Transcriptional Regulation, RIKEN Research Center for Allergy and Immunology, Yokohama, Kanagawa, Japan
| | - Femke van Wijk
- Division of Developmental Immunology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| | - Ryo Shinnakasu
- Division of Developmental Immunology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| | - Yoshinori Naoe
- Laboratory for Transcriptional Regulation, RIKEN Research Center for Allergy and Immunology, Yokohama, Kanagawa, Japan
| | | | - Yujun Huang
- Division of Developmental Immunology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| | - Florence Lambolez
- Division of Developmental Immunology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| | - Michael Docherty
- Division of Developmental Immunology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| | - Antoine Attinger
- Division of Developmental Immunology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| | - Jr-Wen Shui
- Division of Developmental Immunology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| | - Gisen Kim
- Division of Developmental Immunology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| | - Christopher J. Lena
- Division of Developmental Immunology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| | - Shinya Sakaguchi
- Institute of Immunology, Center for Physiology, Pathophysiology and Immunology, Medical University of Vienna Lazarettgasse 19A -1090, Vienna, Austria
| | - Chizuko Miyamoto
- Laboratory for Transcriptional Regulation, RIKEN Research Center for Allergy and Immunology, Yokohama, Kanagawa, Japan
| | - Peng Wang
- Department of Immunology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
- Division of Vaccine Discovery, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| | | | - Yunji Park
- Division of Developmental Immunology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chuo-ku, Chiba 260-8670, Japan
| | | | - Wilfried Ellmeier
- Institute of Immunology, Center for Physiology, Pathophysiology and Immunology, Medical University of Vienna Lazarettgasse 19A -1090, Vienna, Austria
| | - Mitchell Kronenberg
- Division of Developmental Immunology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| | - Ichiro Taniuchi
- Laboratory for Transcriptional Regulation, RIKEN Research Center for Allergy and Immunology, Yokohama, Kanagawa, Japan
| | - Hilde Cheroutre
- Division of Developmental Immunology, La Jolla Institute for Allergy & Immunology, La Jolla, CA 92037, USA
| |
Collapse
|
47
|
Abstract
Sant and McMichael discuss new advances in detecting CD4+ T cells at the right time and place during viral infection. Protective immunity to chronic and acute viral infection relies on both the innate and adaptive immune response. Although neutralizing antibody production by B cells and cytotoxic activity of CD8+ T cells are well-accepted components of the adaptive immune response to viruses, identification of the specific role of CD4+ T cells in protection has been more challenging to establish. Delineating the contribution of CD4+ T cells has been complicated by their functional heterogeneity, breadth in antigen specificity, transient appearance in circulation, and sequestration in tissue sites of infection. In this minireview, we discuss recent progress in identifying the multiple roles of CD4+ T cells in orchestrating and mediating the immune responses against viral pathogens. We highlight several recent reports, including one published in this issue, that have employed comprehensive and sophisticated approaches to provide new evidence for CD4+ T cells as direct effectors in antiviral immunity.
Collapse
Affiliation(s)
- Andrea J Sant
- David H. Smith Center for Vaccine Biology and the Immunology Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14623, USA.
| | | |
Collapse
|
48
|
Illingworth J, Butler NS, Roetynck S, Mwacharo J, Pierce SK, Bejon P, Crompton PD, Marsh K, Ndungu FM. Chronic exposure to Plasmodium falciparum is associated with phenotypic evidence of B and T cell exhaustion. THE JOURNAL OF IMMUNOLOGY 2012; 190:1038-47. [PMID: 23264654 DOI: 10.4049/jimmunol.1202438] [Citation(s) in RCA: 233] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Naturally acquired immunity to malaria develops slowly, requiring several years of repeated exposure to be effective. The cellular and molecular factors underlying this observation are only partially understood. Recent studies suggest that chronic Plasmodium falciparum exposure may induce functional exhaustion of lymphocytes, potentially impeding optimal control of infection. However, it remains unclear whether the "atypical" memory B cells (MBCs) and "exhausted" CD4 T cells described in humans exposed to endemic malaria are driven by P. falciparum per se or by other factors commonly associated with malaria, such as coinfections and malnutrition. To address this critical question we took advantage of a "natural" experiment near Kilifi, Kenya, and compared profiles of B and T cells of children living in a rural community where P. falciparum transmission is ongoing to the profiles of age-matched children living under similar conditions in a nearby community where P. falciparum transmission ceased 5 y prior to this study. We found that continuous exposure to P. falciparum drives the expansion of atypical MBCs. Persistent P. falciparum exposure was associated with an increased frequency of CD4 T cells expressing phenotypic markers of exhaustion, both programmed cell death-1 (PD-1) alone and PD-1 in combination with lymphocyte-activation gene-3 (LAG-3). This expansion of PD-1-expressing and PD-1/LAG-3-coexpressing CD4 T cells was largely confined to CD45RA(+) CD4 T cells. The percentage of CD45RA(+)CD27(+) CD4 T cells coexpressing PD-1 and LAG-3 was inversely correlated with frequencies of activated and classical MBCs. Taken together, these results suggest that P. falciparum infection per se drives the expansion of atypical MBCs and phenotypically exhausted CD4 T cells, which has been reported in other endemic areas.
Collapse
Affiliation(s)
- Joseph Illingworth
- Kenya Medical Research Institute, Centre for Geographical Medicine Research Coast, Kilifi, Kenya
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Antoine P, Olislagers V, Huygens A, Lecomte S, Liesnard C, Donner C, Marchant A. Functional exhaustion of CD4+ T lymphocytes during primary cytomegalovirus infection. THE JOURNAL OF IMMUNOLOGY 2012; 189:2665-72. [PMID: 22865914 DOI: 10.4049/jimmunol.1101165] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Human CMV establishes lifelong persistence after primary infection. Chronic CMV infection is associated with intermittent viral reactivation inducing high frequencies of CD4+ T lymphocytes with potent antiviral and helper properties. Primary CMV infection is characterized by an intense viral replication lasting for several months. The impact of this prolonged exposure to high Ag loads on the functionality of CD4+ T cells remains incompletely understood. In pregnant women with primary CMV infection, we observed that CMV-specific CD4+ T lymphocytes had a decreased capacity to proliferate and to produce IL-2. A very large proportion of CMV-specific CD4+ T cells had downregulated the expression of CD28, a costimulatory molecule centrally involved in the production of IL-2. Unexpectedly, both CD28+ and CD28+ CD4+ T cells produced low levels of IL-2. This defective production of IL-2 was part of a larger downregulation of cytokine production. Indeed, CMV-specific CD4+ T cells produced lower amounts of IFN-γ and TNF-α and showed lower functional avidity during primary as compared with chronic infection. Increased programmed death-1 expression was observed in CD28+ CMV-specific CD4+ T cells, and programmed death-1 inhibition increased proliferative responses. These results indicate that primary CMV infection is associated with the exhaustion of CMV-specific CD4+ T cells displaying low functional avidity for viral Ags.
Collapse
Affiliation(s)
- Pierre Antoine
- Institute for Medical Immunology, Université Libre de Bruxelles, 6041 Charleroi, Belgium
| | | | | | | | | | | | | |
Collapse
|
50
|
Cancer Immunotherapy by Retargeting of Immune Effector Cells via Recombinant Bispecific Antibody Constructs. Antibodies (Basel) 2012. [DOI: 10.3390/antib1020172] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
|