1
|
Cuesta-Martín de la Cámara R, Torices-Pajares A, Miguel-Berenguel L, Reche-Yebra K, Frauca-Remacha E, Hierro-Llanillo L, Muñoz-Bartolo G, Lledín-Barbacho MD, Gutiérrez-Arroyo A, Martínez-Feito A, López-Granados E, Sánchez-Zapardiel E. Epstein-Barr virus-specific T-cell response in pediatric liver transplant recipients: a cross-sectional study by multiparametric flow cytometry. Front Immunol 2024; 15:1479472. [PMID: 39512353 PMCID: PMC11540634 DOI: 10.3389/fimmu.2024.1479472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 10/07/2024] [Indexed: 11/15/2024] Open
Abstract
Background Epstein-Barr virus (EBV) specific T-cell response measurement can help adjust immunosuppression in transplant patients with persistent infections. We aim to define T-cell responses against EBV in a cohort of pediatric liver-transplant patients. Methods Thirty-eight immunosuppressed pediatric liver-transplant patients (IP) and 25 EBV-seropositive healthy-adult controls (HC) were included in our cross-sectional study. Based on their EBV serological (S) and viral load (VL) status, patients were categorized into IP-SNEG, IP-SPOSVLNEG and IP-SPOSVLPOS groups. T-cell response was assessed at two timepoints by stimulating cells with EBV peptides (PepTivator®) and performing intracellular-cytokine and activation-induced marker staining. Background subtraction was used to determine EBV-specific T-lymphocyte frequency. Results Polyfunctional CD8+ T cells indicated previous EBV contact (IP-SNEG 0.00% vs IP-SPOS 0.04% and HC 0.02%; p=0.001 and p=0.01, respectively). Polyfunctional CD8+CD107a+IFNɣ+IL2-TNFα- profile was increased in serology-positive (IP-SNEG 0.01% vs IP-SPOS 0.13% and HC 0.03%; p=0.01 and p=0.50, respectively) and viral-load positive (IP-SPOSVLPOS 0.43% vs IP-SPOSVLNEG 0.07% and HC 0.03%; p=0.03 and p=0.001, respectively) patients. Central-memory cells were increased among serology-positive adults (IP-SNEG 0.00% vs IP-SPOS 0.13% and HC 4.33%; p=0.58 and p=0.002, respectively). At the second timepoint, IP-SNEG patients remained negative (first visit 0.01% vs second visit 0.00%, p=0.44). On the other hand, IP-SPOSVLPOS patients had cleared viral loads and, subsequently, decreased polyfunctional CD8+CD107a+IFNɣ+IL2-TNFα- cells (first visit 0.43% vs second visit 0.10%, p=0.81). Conclusion Polyfunctional CD8+ EBV-specific T-cell response allows detecting EBV previous contact in liver-transplant children. %CD8+CD107a+IFNɣ+IL2-TNFα- is increased in patients with positive viral loads. Central memory CD4+ T-cell population more effectively determines prior EBV-exposure in adults.
Collapse
Affiliation(s)
- Ricardo Cuesta-Martín de la Cámara
- Clinical Immunology Department, University Hospital La Paz, Madrid, Spain
- Lymphocyte Pathophysiology in Immunodeficiencies Group, La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
- Medicine and surgery Department, Autonomous University of Madrid, Madrid, Spain
| | - Andrea Torices-Pajares
- Lymphocyte Pathophysiology in Immunodeficiencies Group, La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | | | - Keren Reche-Yebra
- Lymphocyte Pathophysiology in Immunodeficiencies Group, La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Esteban Frauca-Remacha
- Paediatric Hepatology Department, University Hospital La Paz, Madrid, Spain
- European Reference Network (ERN) RARE LIVER, Madrid, Spain
- European Reference Network (ERN) TransplantChild, Madrid, Spain
| | - Loreto Hierro-Llanillo
- Paediatric Hepatology Department, University Hospital La Paz, Madrid, Spain
- European Reference Network (ERN) RARE LIVER, Madrid, Spain
- European Reference Network (ERN) TransplantChild, Madrid, Spain
| | - Gema Muñoz-Bartolo
- Paediatric Hepatology Department, University Hospital La Paz, Madrid, Spain
- European Reference Network (ERN) RARE LIVER, Madrid, Spain
- European Reference Network (ERN) TransplantChild, Madrid, Spain
| | - María Dolores Lledín-Barbacho
- Paediatric Hepatology Department, University Hospital La Paz, Madrid, Spain
- European Reference Network (ERN) RARE LIVER, Madrid, Spain
- European Reference Network (ERN) TransplantChild, Madrid, Spain
| | | | - Ana Martínez-Feito
- Clinical Immunology Department, University Hospital La Paz, Madrid, Spain
| | - Eduardo López-Granados
- Clinical Immunology Department, University Hospital La Paz, Madrid, Spain
- Lymphocyte Pathophysiology in Immunodeficiencies Group, La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
- European Reference Network (ERN) TransplantChild, Madrid, Spain
- Centre for Biomedical Network Research on rare diseases (CIBERER U767), Madrid, Spain
| | - Elena Sánchez-Zapardiel
- Clinical Immunology Department, University Hospital La Paz, Madrid, Spain
- Lymphocyte Pathophysiology in Immunodeficiencies Group, La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
- European Reference Network (ERN) TransplantChild, Madrid, Spain
| |
Collapse
|
2
|
Zhao Y, Cai C, Samir J, Palgen JL, Keoshkerian E, Li H, Bull RA, Luciani F, An H, Lloyd AR. Human CD8 T-stem cell memory subsets phenotypic and functional characterization are defined by expression of CD122 or CXCR3. Eur J Immunol 2021; 51:1732-1747. [PMID: 33844287 DOI: 10.1002/eji.202049057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/07/2021] [Accepted: 04/07/2021] [Indexed: 11/12/2022]
Abstract
Long-lived T-memory stem cells (TSCM ) are key to both naturally occurring and vaccine-conferred protection against infection. These cells are characterized by the CD45RA+ CCR7+ CD95+ phenotype. Significant heterogeneity within the TSCM population is recognized, but distinguishing surface markers and functional characterization of potential subsets are lacking. Human CD8 TSCM subsets were identified in healthy subjects who had been previously exposed to CMV or Influenza (Flu) virus in flow cytometry by expression of CD122 or CXCR3, and then characterized in proliferation, multipotency, self-renewal, and intracellular cytokine production (TNF-α, IL-2, IFN-γ), together with transcriptomic profiles. The TSCM CD122hi -expressing subset (versus CD122lo ) demonstrated greater proliferation, greater multipotency, and enhanced polyfunctionality with higher frequencies of triple positive (TNF-α, IL-2, IFN-γ) cytokine-producing cells upon exposure to recall antigen. The TSCM CXCR3lo subpopulation also had increased proliferation and polyfunctional cytokine production. Transcriptomic analysis further showed that the TSCM CD122hi population had increased expression of activation and homing molecules, such as Ccr6, Cxcr6, Il12rb, and Il18rap, and downregulated cell proliferation inhibitors, S100A8 and S100A9. These data reveal that the TSCM CD122hi phenotype is associated with increased proliferation, enhanced multipotency and polyfunctionality with an activated memory-cell like transcriptional profile, and hence, may be favored for induction by immunization and for adoptive immunotherapy.
Collapse
Affiliation(s)
- Yanran Zhao
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Curtis Cai
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Jerome Samir
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Jean-Louis Palgen
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Elizabeth Keoshkerian
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Hui Li
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Rowena A Bull
- School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Fabio Luciani
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia.,School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Hongyan An
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| | - Andrew R Lloyd
- Viral Immunology Systems Program (VISP), The Kirby Institute, University of New South Wales, Sydney, Australia
| |
Collapse
|
3
|
Helmold Hait S, Hogge CJ, Rahman MA, Hunegnaw R, Mushtaq Z, Hoang T, Robert-Guroff M. T FH Cells Induced by Vaccination and Following SIV Challenge Support Env-Specific Humoral Immunity in the Rectal-Genital Tract and Circulation of Female Rhesus Macaques. Front Immunol 2021; 11:608003. [PMID: 33584682 PMCID: PMC7876074 DOI: 10.3389/fimmu.2020.608003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 12/07/2020] [Indexed: 11/13/2022] Open
Abstract
T follicular helper (TFH) cells are pivotal in lymph node (LN) germinal center (GC) B cell affinity maturation. Circulating CXCR5+ CD4+ T (cTFH) cells have supported memory B cell activation and broadly neutralizing antibodies in HIV controllers. We investigated the contribution of LN SIV-specific TFH and cTFH cells to Env-specific humoral immunity in female rhesus macaques following a mucosal Ad5hr-SIV recombinant priming and SIV gp120 intramuscular boosting vaccine regimen and following SIV vaginal challenge. TFH and B cells were characterized by flow cytometry. B cell help was evaluated in TFH-B cell co-cultures and by real-time PCR. Vaccination induced Env-specific TFH and Env-specific memory (ESM) B cells in LNs. LN Env-specific TFH cells post-priming and GC ESM B cells post-boosting correlated with rectal Env-specific IgA titers, and GC B cells at the same timepoints correlated with vaginal Env-specific IgG titers. Vaccination expanded cTFH cell responses, including CD25+ Env-specific cTFH cells that correlated negatively with vaginal Env-specific IgG titers but positively with rectal Env-specific IgA titers. Although cTFH cells post-2nd boost positively correlated with viral-loads following SIV challenge, cTFH cells of SIV-infected and protected macaques supported maturation of circulating B cells into plasma cells and IgA release in co-culture. Additionally, cTFH cells of naïve macaques promoted upregulation of genes associated with B cell proliferation, BCR engagement, plasma cell maturation, and antibody production, highlighting the role of cTFH cells in blood B cell maturation. Vaccine-induced LN TFH and GC B cells supported anti-viral mucosal immunity while cTFH cells provided B cell help in the periphery during immunization and after SIV challenge. Induction of TFH responses in blood and secondary lymphoid organs is likely desirable for protective efficacy of HIV vaccines.
Collapse
Affiliation(s)
- Sabrina Helmold Hait
- Immune Biology of Retroviral Infection Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Christopher James Hogge
- Immune Biology of Retroviral Infection Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Mohammad Arif Rahman
- Immune Biology of Retroviral Infection Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Ruth Hunegnaw
- Immune Biology of Retroviral Infection Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Zuena Mushtaq
- Immune Biology of Retroviral Infection Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Tanya Hoang
- Immune Biology of Retroviral Infection Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Marjorie Robert-Guroff
- Immune Biology of Retroviral Infection Section, Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
4
|
CD73 + CD127 high Long-Term Memory CD4 T Cells Are Highly Proliferative in Response to Recall Antigens and Are Early Targets in HIV-1 Infection. Int J Mol Sci 2021; 22:ijms22020912. [PMID: 33477692 PMCID: PMC7831934 DOI: 10.3390/ijms22020912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 11/17/2022] Open
Abstract
HIV-1 infection rapidly leads to a loss of the proliferative response of memory CD4+ T lymphocytes, when cultured with recall antigens. We report here that CD73 expression defines a subset of resting memory CD4+ T cells in peripheral blood, which highly express the α-chain of the IL-7 receptor (CD127), but not CD38 or Ki-67, yet are highly proliferative in response to mitogen and recall antigens, and to IL-7, in vitro. These cells also preferentially express CCR5 and produce IL-2. We reasoned that CD73+ memory CD4+ T cells decrease very early in HIV-1 infection. Indeed, CD73+ memory CD4+ T cells comprised a median of 7.5% (interquartile range: 4.5-10.4%) of CD4+ T cells in peripheral blood from healthy adults, but were decreased in primary HIV-1 infection to a median of 3.7% (IQR: 2.6-6.4%; p = 0.002); and in chronic HIV-1 infection to 1.9% (IQR: 1.1-3%; p < 0.0001), and were not restored by antiretroviral therapy. Moreover, we found that a significant proportion of CD73+ memory CD4+ T cells were skewed to a gut-homing phenotype, expressing integrins α4 and β7, CXCR3, CCR6, CD161 and CD26. Accordingly, 20% of CD4+ T cells present in gut biopsies were CD73+. In HIV+ subjects, purified CD73+ resting memory CD4+ T cells in PBMC were infected with HIV-1 DNA, determined by real-time PCR, to the same level as for purified CD73-negative CD4+ T cells, both in untreated and treated subjects. Therefore, the proliferative CD73+ subset of memory CD4+ T cells is disproportionately reduced in HIV-1 infection, but, unexpectedly, their IL-7 dependent long-term resting phenotype suggests that residual infected cells in this subset may contribute significantly to the very long-lived HIV proviral DNA reservoir in treated subjects.
Collapse
|
5
|
Activation-induced surface proteins in the identification of antigen-responsive CD4 T cells. Immunol Lett 2019; 219:1-7. [PMID: 31881234 DOI: 10.1016/j.imlet.2019.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 12/09/2019] [Accepted: 12/22/2019] [Indexed: 02/06/2023]
Abstract
Identification of antigen specificity of CD4 T cells is instrumental in understanding adaptive immune responses in health and disease. The high diversity of CD4 T cell repertoire combined with the functional heterogeneity of the compartment poses a challenge to the assessment of CD4 T cell responses. In spite of that, multiple technologies allow direct or indirect interrogation of antigen specificity of CD4 T cells. In the last decade, multiple surface proteins have been established as cytokine-independent surrogates of in vitro CD4 T cell activation, and have found applications in the live identification and isolation of antigen-responsive CD4 T cells. Here we review the current knowledge of the surface proteins that permit identification of viable antigen-responsive CD4 T cells with high specificity, including those capable of identifying specialized CD4 T subsets such as germinal center follicular helper T cells and CD4 regulatory T cells.
Collapse
|
6
|
Jiang W, Wragg KM, Tan HX, Kelly HG, Wheatley AK, Kent SJ, Juno JA. Identification of murine antigen-specific T follicular helper cells using an activation-induced marker assay. J Immunol Methods 2019; 467:48-57. [DOI: 10.1016/j.jim.2019.02.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 01/04/2023]
|
7
|
Early T Follicular Helper Cell Responses and Germinal Center Reactions Are Associated with Viremia Control in Immunized Rhesus Macaques. J Virol 2019; 93:JVI.01687-18. [PMID: 30463978 DOI: 10.1128/jvi.01687-18] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 11/13/2018] [Indexed: 12/15/2022] Open
Abstract
T follicular helper (TFH) cells are fundamental in germinal center (GC) maturation and selection of antigen-specific B cells within secondary lymphoid organs. GC-resident TFH cells have been fully characterized in human immunodeficiency virus (HIV) infection. However, the role of GC TFH cells in GC B cell responses following various simian immunodeficiency virus (SIV) vaccine regimens in rhesus macaques (RMs) has not been fully investigated. We characterized GC TFH cells of RMs over the course of a mucosal/systemic vaccination regimen to elucidate GC formation and SIV humoral response generation. Animals were mucosally primed twice with replicating adenovirus type 5 host range mutant (Ad5hr)-SIV recombinants and systemically boosted with ALVAC-SIVM766Gag/Pro/gp120-TM and SIVM766&CG7V gD-gp120 proteins formulated in alum hydroxide (ALVAC/Env) or DNA encoding SIVenv/SIVGag/rhesus interleukin 12 (IL-12) plus SIVM766&CG7V gD-gp120 proteins formulated in alum phosphate (DNA&Env). Lymph nodes were biopsied in macaque subgroups prevaccination and at day 3, 7, or 14 after the 2nd Ad5hr-SIV prime and the 2nd vector/Env boost. Evaluations of GC TFH and GC B cell dynamics including correlation analyses supported a significant role for early GC TFH cells in providing B cell help during initial phases of GC formation. GC TFH responses at day 3 post-mucosal priming were consistent with generation of Env-specific memory B cells in GCs and elicitation of prolonged Env-specific humoral immunity in the rectal mucosa. GC Env-specific memory B cell responses elicited early post-systemic boosting correlated significantly with decreased viremia postinfection. Our results highlight the importance of early GC TFH cell responses for robust GC maturation and generation of long-lasting SIV-specific humoral responses at mucosal and systemic sites. Further investigation of GC TFH cell dynamics should facilitate development of an efficacious HIV vaccine.IMPORTANCE The modest HIV protection observed in the human RV144 vaccine trial associated antibody responses with vaccine efficacy. T follicular helper (TFH) cells are CD4+ T cells that select antibody secreting cells with high antigenic affinity in germinal centers (GCs) within secondary lymphoid organs. To evaluate the role of TFH cells in eliciting prolonged virus-specific humoral responses, we vaccinated rhesus macaques with a combined mucosal prime/systemic boost regimen followed by repeated low-dose intrarectal challenges with SIV, mimicking human exposure to HIV-1. Although the vaccine regimen did not prevent SIV infection, decreased viremia was observed in the immunized macaques. Importantly, vaccine-induced TFH responses elicited at day 3 postimmunization and robust GC maturation were strongly associated. Further, early TFH-dependent SIV-specific B cell responses were also correlated with decreased viremia. Our findings highlight the contribution of early vaccine-induced GC TFH responses to elicitation of SIV-specific humoral immunity and implicate their participation in SIV control.
Collapse
|
8
|
Gaido CM, Granland C, Laing IA, Souëf PNL, Thomas WR, Currie AJ, Hales BJ. T-cell responses against rhinovirus species A and C in asthmatic and healthy children. IMMUNITY INFLAMMATION AND DISEASE 2017; 6:143-153. [PMID: 29124902 PMCID: PMC5818445 DOI: 10.1002/iid3.206] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/13/2017] [Accepted: 10/18/2017] [Indexed: 12/28/2022]
Abstract
Background Infections by rhinovirus (RV) species A and C are the most common causes of exacerbations of asthma and a major cause of exacerbations of other acute and chronic respiratory diseases. Infections by both species are prevalent in pre‐school and school‐aged children and, particularly for RV‐C, can cause severe symptoms and a need for hospitalization. While associations between RV infection and asthma are well established, the adaptive immune‐mechanisms by which RV infections influence asthma exacerbations are yet to be defined. Objective The aim of this study was to characterize and compare T‐cell responses between RV‐A and RV‐C and to test the hypothesis that T‐cell responses would differ between asthmatic children and healthy controls. Methods A multi‐parameter flow cytometry assay was used to characterize the in vitro recall T‐cell response against RV‐A and RV‐C in PBMCs from children with acute asthma (n = 22) and controls (n = 26). The responses were induced by pools of peptides containing species‐specific VP1 epitopes of RV‐A and RV‐C. Results Regardless of children's clinical status, all children that responded to the in vitro stimulation (>90%) had a similar magnitude of CD4+ T‐cell responses to RV‐A and RV‐C. However, asthmatic children had a significantly lower number of circulating regulatory T cells (Tregs), and healthy controls had significantly more Tregs induced by RV‐A than RV‐C. Conclusions and Clinical Relevance The comparable recall memory T‐cell responses in asthmatic and control children to both RV‐A and RV‐C show that differences in the antibody and inflammatory responses previously described are likely to be due to regulation, with a demonstrated candidate being reduced regulatory T‐cells. The reduced Treg numbers demonstrated here could explain the asthmatic's inability to appropriately control immunopathological responses to RV infections.
Collapse
Affiliation(s)
- Cibele M Gaido
- Telethon Kids Institute, The University of Western Australia, Perth, Australia.,School of Paediatrics and Child Health, The University of Western Australia, Perth, Australia
| | - Caitlyn Granland
- Telethon Kids Institute, The University of Western Australia, Perth, Australia.,School of Paediatrics and Child Health, The University of Western Australia, Perth, Australia
| | - Ingrid A Laing
- Telethon Kids Institute, The University of Western Australia, Perth, Australia.,School of Paediatrics and Child Health, The University of Western Australia, Perth, Australia
| | - Peter N Le Souëf
- School of Paediatrics and Child Health, The University of Western Australia, Perth, Australia.,Princess Margaret Hospital for Children, Perth, Australia
| | - Wayne R Thomas
- Telethon Kids Institute, The University of Western Australia, Perth, Australia
| | - Andrew J Currie
- School of Paediatrics and Child Health, The University of Western Australia, Perth, Australia.,School of Veterinary & Life Sciences, Murdoch University, Perth, Australia
| | - Belinda J Hales
- Telethon Kids Institute, The University of Western Australia, Perth, Australia
| |
Collapse
|
9
|
Abstract
Measuring regulatory T cell suppression provides important insight into T cell dysfunction in autoimmune disease. However, to date, suppression assays are limited by the requirement for freshly isolated cells, and significant cell numbers. Here, we present a novel and rapid in vitro assay using effector T cell surface expression of both CD25 and CD134 as a surrogate marker of regulatory T cell-mediated suppression. This surface marker-based suppression assay works for frozen samples and for samples with limited cell numbers. It is also shorter taking two days to complete compared to the four days required for proliferation-based assays. Furthermore, this assay works with both in vitro expanded and natural Tregs, as well as anti-CD3/anti-CD28 bead-based and APC stimulation conditions. In conclusion, we have developed and validated a new suppression assay for cryopreserved samples with limited cell numbers that may be helpful to investigate T cell regulation in the context of infection or autoimmune diseases.
Collapse
|
10
|
Cook L, Munier CML, Seddiki N, van Bockel D, Ontiveros N, Hardy MY, Gillies JK, Levings MK, Reid HH, Petersen J, Rossjohn J, Anderson RP, Zaunders JJ, Tye-Din JA, Kelleher AD. Circulating gluten-specific FOXP3 +CD39 + regulatory T cells have impaired suppressive function in patients with celiac disease. J Allergy Clin Immunol 2017; 140:1592-1603.e8. [PMID: 28283419 DOI: 10.1016/j.jaci.2017.02.015] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 02/03/2017] [Accepted: 02/16/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Celiac disease is a chronic immune-mediated inflammatory disorder of the gut triggered by dietary gluten. Although the effector T-cell response in patients with celiac disease has been well characterized, the role of regulatory T (Treg) cells in the loss of tolerance to gluten remains poorly understood. OBJECTIVE We sought to define whether patients with celiac disease have a dysfunction or lack of gluten-specific forkhead box protein 3 (FOXP3)+ Treg cells. METHODS Treated patients with celiac disease underwent oral wheat challenge to stimulate recirculation of gluten-specific T cells. Peripheral blood was collected before and after challenge. To comprehensively measure the gluten-specific CD4+ T-cell response, we paired traditional IFN-γ ELISpot with an assay to detect antigen-specific CD4+ T cells that does not rely on tetramers, antigen-stimulated cytokine production, or proliferation but rather on antigen-induced coexpression of CD25 and OX40 (CD134). RESULTS Numbers of circulating gluten-specific Treg cells and effector T cells both increased significantly after oral wheat challenge, peaking at day 6. Surprisingly, we found that approximately 80% of the ex vivo circulating gluten-specific CD4+ T cells were FOXP3+CD39+ Treg cells, which reside within the pool of memory CD4+CD25+CD127lowCD45RO+ Treg cells. Although we observed normal suppressive function in peripheral polyclonal Treg cells from patients with celiac disease, after a short in vitro expansion, the gluten-specific FOXP3+CD39+ Treg cells exhibited significantly reduced suppressive function compared with polyclonal Treg cells. CONCLUSION This study provides the first estimation of FOXP3+CD39+ Treg cell frequency within circulating gluten-specific CD4+ T cells after oral gluten challenge of patients with celiac disease. FOXP3+CD39+ Treg cells comprised a major proportion of all circulating gluten-specific CD4+ T cells but had impaired suppressive function, indicating that Treg cell dysfunction might be a key contributor to disease pathogenesis.
Collapse
Affiliation(s)
- Laura Cook
- Immunovirology and Pathogenesis Program, The Kirby Institute, UNSW Sydney, Sydney, Australia; St Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Sydney, Australia.
| | - C Mee Ling Munier
- Immunovirology and Pathogenesis Program, The Kirby Institute, UNSW Sydney, Sydney, Australia
| | - Nabila Seddiki
- Immunovirology and Pathogenesis Program, The Kirby Institute, UNSW Sydney, Sydney, Australia; St Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Sydney, Australia
| | - David van Bockel
- Immunovirology and Pathogenesis Program, The Kirby Institute, UNSW Sydney, Sydney, Australia
| | - Noé Ontiveros
- Immunology Division, Walter and Eliza Hall Institute, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Melinda Y Hardy
- Immunology Division, Walter and Eliza Hall Institute, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Jana K Gillies
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Megan K Levings
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hugh H Reid
- Infection and Immunity Program, The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Australia; Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Australia
| | - Jan Petersen
- Infection and Immunity Program, The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Australia; Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program, The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Australia; Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Monash University, Clayton, Australia; Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, United Kingdom
| | - Robert P Anderson
- Immunology Division, Walter and Eliza Hall Institute, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia; ImmusanT, Cambridge, Mass
| | - John J Zaunders
- Immunovirology and Pathogenesis Program, The Kirby Institute, UNSW Sydney, Sydney, Australia; St Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Sydney, Australia
| | - Jason A Tye-Din
- Immunology Division, Walter and Eliza Hall Institute, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia; Department of Gastroenterology, Royal Melbourne Hospital, Parkville, Australia
| | - Anthony D Kelleher
- Immunovirology and Pathogenesis Program, The Kirby Institute, UNSW Sydney, Sydney, Australia; St Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Sydney, Australia
| |
Collapse
|
11
|
Keoshkerian E, Hunter M, Cameron B, Nguyen N, Sugden P, Bull R, Zekry A, Maher L, Seddiki N, Zaunders J, Kelleher A, Lloyd AR. Hepatitis C-specific effector and regulatory CD4 T-cell responses are associated with the outcomes of primary infection. J Viral Hepat 2016; 23:985-993. [PMID: 27558465 DOI: 10.1111/jvh.12576] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 06/29/2016] [Indexed: 12/31/2022]
Abstract
Clearance of primary hepatitis C virus (HCV) infection has been associated with strong and broadly targeted cellular immune responses. This study aimed to characterize HCV-specific CD4+ effector and regulatory T-cell numbers and cytokine production during primary infection. Antigen-specific CD4+ T-cell responses were investigated in a longitudinal cohort of subjects from pre-infection to postoutcome, including subjects who cleared [n=12] or became chronically infected [n=17]. A cross-sectional cohort with previously cleared, or chronic infection [n=15 for each], was also studied. Peripheral blood mononuclear cells were incubated with HCV antigens and surface stained for T-effector (CD4+CD25high CD134+CD39-) and T-regulatory (CD4+CD25high CD134+CD39+) markers, and culture supernatants assayed for cytokine production. Contrary to expectations, the breadth and magnitude of the HCV-specific CD4+ T-cell responses were higher in subjects who became chronically infected. Subjects who cleared the virus had HCV-specific CD4+ T-cell responses dominated by effector T cells and produced higher levels of IFN-γ, in contrast to HCV-specific CD4+ T-cell responses dominated by regulatory T cells and more IL-10 production in those who became chronically infected. Better understanding of the role of antigen-specific CD4+ T-cell responses in primary HCV will further define pathogenesis and help guide development of a preventative vaccine.
Collapse
Affiliation(s)
- E Keoshkerian
- UNSW Australia, Kirby Institute (Viral Immunology Systems Program, VISP) and School of Medical Sciences (SOMS), Kensington, NSW, Australia
| | - M Hunter
- UNSW Australia, SOMS (Infection and Immunology Research Centre, IIRC), Kensington, NSW, Australia
| | - B Cameron
- UNSW Australia, SOMS (Infection and Immunology Research Centre, IIRC), Kensington, NSW, Australia
| | - N Nguyen
- UNSW Australia, SOMS (Infection and Immunology Research Centre, IIRC), Kensington, NSW, Australia
| | - P Sugden
- UNSW Australia, SOMS (Infection and Immunology Research Centre, IIRC), Kensington, NSW, Australia
| | - R Bull
- UNSW Australia, Kirby Institute (Viral Immunology Systems Program, VISP) and School of Medical Sciences (SOMS), Kensington, NSW, Australia
| | - A Zekry
- UNSW Australia, St George and Sutherland Clinical School, Sydney, NSW, Australia
| | - L Maher
- UNSW Australia, Kirby Institute (Viral Hepatitis Epidemiology and Prevention Program VHEPP), Kensington, NSW, Australia
| | - N Seddiki
- The Vaccine Research Institute (VRI), INSERM, Créteil, France
| | - J Zaunders
- UNSW Australia, Kirby Institute (Immunovirology and Pathogenesis Program, IVPP), Kensington, NSW, Australia
| | - A Kelleher
- UNSW Australia, Kirby Institute (Immunovirology and Pathogenesis Program, IVPP), Kensington, NSW, Australia
| | - A R Lloyd
- UNSW Australia, Kirby Institute (Viral Immunology Systems Program, VISP) and School of Medical Sciences (SOMS), Kensington, NSW, Australia
| | | |
Collapse
|
12
|
Fouad H, El Raziky M, Hassan EM, Aziz GMA, Darweesh SK, Sayed AR. Regulatory and activated effector T cells in chronic hepatitis C virus: Relation to autoimmunity. World J Hepatol 2016; 8:1287-1294. [PMID: 27843539 PMCID: PMC5084058 DOI: 10.4254/wjh.v8.i30.1287] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 07/30/2016] [Accepted: 08/27/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate how Tregs are regulated in chronic hepatitis C virus (HCV) patients via assessment of Tregs markers (granzyme 2, CD69 and FoxP3), Teffs markers [TNFRSF4 (OX40), INFG] and CD4, CD25 genes. METHODS A prospective study was conducted on 120 subjects divided into 4 groups: Group I (n = 30) treatment naïve chronic HCV patients; Group II (n = 30) chronic HCV treated with Peg/Riba; Group III (n = 30) chronic HCV associated with non-organ specific autoantibody and Group IV (n = 30) healthy persons as a control group. Tregs and Teffs markers were assessed in peripheral blood mononuclear cells by quantitative real time reverse transcriptase-polymerase chain reaction. RESULTS Chronic HCV patients exhibited significant higher levels of both Teffs and Tregs in comparison to healthy control group. Tregs markers were significantly decreased in Peg/Riba treated HCV patients in comparison to treatment naïve HCV group. In HCV patients with antinuclear antibody (ANA) +ve, Tregs markers were significantly decreased in comparison to all other studied groups. Teffs markers were significantly elevated in all HCV groups in comparison to control and in HCV group with ANA +ve in comparison to treatment naïve HCV group. CONCLUSION Elevated Tregs cells in chronic HCV patients dampen both CD4+ and CD8+ autologous T cell immune response. Interferon-α and ribavirin therapy suppress proliferation of Tregs. More significant suppression of Tregs was observed in HCV patients with autoantibodies favoring pathological autoimmune response.
Collapse
Affiliation(s)
- Hanan Fouad
- Hanan Fouad, Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
| | - Maissa El Raziky
- Hanan Fouad, Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
| | - Eman Medhat Hassan
- Hanan Fouad, Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
| | - Ghada Mahmoud Abdel Aziz
- Hanan Fouad, Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
| | - Samar K Darweesh
- Hanan Fouad, Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
| | - Ahmed Reda Sayed
- Hanan Fouad, Department of Medical Biochemistry, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
13
|
Klaewsongkram J, Thantiworasit P, Sodsai P, Buranapraditkun S, Mongkolpathumrat P. Slow desensitization of imatinib-induced nonimmediate reactions and dynamic changes of drug-specific CD4 +CD25 +CD134 + lymphocytes. Ann Allergy Asthma Immunol 2016; 117:514-519. [PMID: 27788881 DOI: 10.1016/j.anai.2016.08.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 07/22/2016] [Accepted: 08/30/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND Imatinib is a tyrosine kinase inhibitor indicated for the treatment of gastrointestinal stromal tumors (GISTs) and certain neoplastic diseases; however, nonimmediate adverse reactions are common. OBJECTIVE To describe the process of imatinib slow desensitization in patients who experienced nonimmediate reactions to imatinib and the dynamic change in drug-specific CD4+CD25+CD134+ T-lymphocyte percentages. METHODS Five patients diagnosed as having GISTs and with a recent history of imatinib-induced nonimmediate reactions (maculopapular exanthema with eosinophilia, exfoliative dermatitis, palmar-plantar erythrodysesthesia, and drug rash with eosinophilia and systemic symptoms) were desensitized using a slow desensitization protocol. The reintroduced imatinib dosage was stepped up every week starting from 10 mg/d and increasing to 25, 50, 75, 100, 150, 200, and 300 mg/d until the target dose of 400 mg/d was achieved. Prednisolone of up to 30 mg/d was allowed if allergic reactions recurred. The percentages of CD4+CD25+CD134+ T cells present after incubating peripheral blood mononuclear cells with imatinib, at baseline and after successful desensitization, were analyzed using flow cytometric analysis. RESULTS By using a slow desensitization technique, all patients were able to receive 400 mg/d of imatinib, and prednisolone was gradually tapered off. The percentages of imatinib-induced CD4+CD25+CD134+ T cells decreased from a mean (SD) of 11.3% (6.5%) and 13.4% (7.3%) at baseline to 3.2% (0.7%) and 3.0% (1.1%) after successful desensitization, when stimulating peripheral blood mononuclear cells with 1 and 2 μM of imatinib, respectively. CONCLUSION Slow desensitization is a helpful procedure in treating patients with imatinib-induced nonimmediate reactions other than simple maculopapular exanthema. The reduced percentages of imatinib-induced CD4+CD25+CD134+ T cells in these patients may be associated with immune tolerance.
Collapse
Affiliation(s)
- Jettanong Klaewsongkram
- Division of Allergy and Clinical Immunology, Department of Medicine, Faculty of Medicine, and Chulalongkorn Allergy and Clinical Immunology Research Group, Chulalongkorn University, Bangkok, Thailand; King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand.
| | - Pattarawat Thantiworasit
- Division of Allergy and Clinical Immunology, Department of Medicine, Faculty of Medicine, and Chulalongkorn Allergy and Clinical Immunology Research Group, Chulalongkorn University, Bangkok, Thailand
| | - Pimpayao Sodsai
- Center of Excellence in Immunology and Immune-Mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Supranee Buranapraditkun
- Division of Allergy and Clinical Immunology, Department of Medicine, Faculty of Medicine, and Chulalongkorn Allergy and Clinical Immunology Research Group, Chulalongkorn University, Bangkok, Thailand
| | | |
Collapse
|
14
|
Dan JM, Havenar-Daughton C, Silvestri G, Sette A, Crotty S. Response to Comment on “A Cytokine-Independent Approach To Identify Antigen-Specific Human Germinal Center T Follicular Helper Cells and Rare Antigen-Specific CD4+ T Cells in Blood”. THE JOURNAL OF IMMUNOLOGY 2016; 197:2558. [DOI: 10.4049/jimmunol.1601321] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
15
|
Dan JM, Lindestam Arlehamn CS, Weiskopf D, da Silva Antunes R, Havenar-Daughton C, Reiss SM, Brigger M, Bothwell M, Sette A, Crotty S. A Cytokine-Independent Approach To Identify Antigen-Specific Human Germinal Center T Follicular Helper Cells and Rare Antigen-Specific CD4+ T Cells in Blood. THE JOURNAL OF IMMUNOLOGY 2016; 197:983-93. [PMID: 27342848 DOI: 10.4049/jimmunol.1600318] [Citation(s) in RCA: 210] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 05/19/2016] [Indexed: 12/13/2022]
Abstract
Detection of Ag-specific CD4(+) T cells is central to the study of many human infectious diseases, vaccines, and autoimmune diseases. However, such cells are generally rare and heterogeneous in their cytokine profiles. Identification of Ag-specific germinal center (GC) T follicular helper (Tfh) cells by cytokine production has been particularly problematic. The function of a GC Tfh cell is to selectively help adjacent GC B cells via cognate interaction; thus, GC Tfh cells may be stingy cytokine producers, fundamentally different from Th1 or Th17 cells in the quantities of cytokines produced. Conventional identification of Ag-specific cells by intracellular cytokine staining relies on the ability of the CD4(+) T cell to generate substantial amounts of cytokine. To address this problem, we have developed a cytokine-independent activation-induced marker (AIM) methodology to identify Ag-specific GC Tfh cells in human lymphoid tissue. Whereas Group A Streptococcus-specific GC Tfh cells produced minimal detectable cytokines by intracellular cytokine staining, the AIM method identified 85-fold more Ag-specific GC Tfh cells. Intriguingly, these GC Tfh cells consistently expressed programmed death ligand 1 upon activation. AIM also detected non-Tfh cells in lymphoid tissue. As such, we applied AIM for identification of rare Ag-specific CD4(+) T cells in human peripheral blood. Dengue, tuberculosis, and pertussis vaccine-specific CD4(+) T cells were readily detectable by AIM. In summary, cytokine assays missed 98% of Ag-specific human GC Tfh cells, reflecting the biology of these cells, which could instead be sensitively identified by coexpression of TCR-dependent activation markers.
Collapse
Affiliation(s)
- Jennifer M Dan
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; Division of Infectious Diseases, University of California, San Diego, La Jolla, CA 92093
| | | | - Daniela Weiskopf
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | | | - Colin Havenar-Daughton
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, La Jolla, CA 92037; and
| | - Samantha M Reiss
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, La Jolla, CA 92037; and
| | | | | | - Alessandro Sette
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037
| | - Shane Crotty
- La Jolla Institute for Allergy and Immunology, La Jolla, CA 92037; Division of Infectious Diseases, University of California, San Diego, La Jolla, CA 92093; Center for HIV/AIDS Vaccine Immunology and Immunogen Discovery, La Jolla, CA 92037; and
| |
Collapse
|
16
|
Tao B, Bernardo K, Eldi P, Chegeni N, Wiese M, Colella A, Kral A, Hayball J, Smith W, Forsyth K, Chataway T. Extended boiling of peanut progressively reduces IgE allergenicity while retaining T cell reactivity. Clin Exp Allergy 2016; 46:1004-14. [DOI: 10.1111/cea.12740] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/09/2016] [Accepted: 03/31/2016] [Indexed: 11/30/2022]
Affiliation(s)
- B. Tao
- Department of Paediatrics and Child Health; Flinders University of South Australia; Adelaide SA Australia
- Flinders Proteomics Facility; Department of Human Physiology; Flinders University of South Australia; Adelaide SA Australia
| | - K. Bernardo
- Flinders Proteomics Facility; Department of Human Physiology; Flinders University of South Australia; Adelaide SA Australia
| | - P. Eldi
- School of Pharmacy and Medical Sciences; University of South Australia; Adelaide SA Australia
| | - N. Chegeni
- Flinders Proteomics Facility; Department of Human Physiology; Flinders University of South Australia; Adelaide SA Australia
| | - M. Wiese
- School of Pharmacy and Medical Sciences; University of South Australia; Adelaide SA Australia
| | - A. Colella
- Department of Immunology; Flinders University of South Australia; Adelaide SA Australia
| | - A. Kral
- Department and School of Medicine; University of Adelaide; Adelaide SA Australia
| | - J. Hayball
- School of Pharmacy and Medical Sciences; University of South Australia; Adelaide SA Australia
| | - W. Smith
- Department and School of Medicine; University of Adelaide; Adelaide SA Australia
| | - K. Forsyth
- Department of Paediatrics and Child Health; Flinders University of South Australia; Adelaide SA Australia
| | - T. Chataway
- Flinders Proteomics Facility; Department of Human Physiology; Flinders University of South Australia; Adelaide SA Australia
| |
Collapse
|
17
|
Phetsouphanh C, Zaunders JJ, Kelleher AD. Detecting Antigen-Specific T Cell Responses: From Bulk Populations to Single Cells. Int J Mol Sci 2015; 16:18878-93. [PMID: 26274954 PMCID: PMC4581277 DOI: 10.3390/ijms160818878] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 07/29/2015] [Accepted: 08/03/2015] [Indexed: 12/18/2022] Open
Abstract
A new generation of sensitive T cell-based assays facilitates the direct quantitation and characterization of antigen-specific T cell responses. Single-cell analyses have focused on measuring the quality and breadth of a response. Accumulating data from these studies demonstrate that there is considerable, previously-unrecognized, heterogeneity. Standard assays, such as the ICS, are often insufficient for characterization of rare subsets of cells. Enhanced flow cytometry with imaging capabilities enables the determination of cell morphology, as well as the spatial localization of the protein molecules within a single cell. Advances in both microfluidics and digital PCR have improved the efficiency of single-cell sorting and allowed multiplexed gene detection at the single-cell level. Delving further into the transcriptome of single-cells using RNA-seq is likely to reveal the fine-specificity of cellular events such as alternative splicing (i.e., splice variants) and allele-specific expression, and will also define the roles of new genes. Finally, detailed analysis of clonally related antigen-specific T cells using single-cell TCR RNA-seq will provide information on pathways of differentiation of memory T cells. With these state of the art technologies the transcriptomics and genomics of Ag-specific T cells can be more definitively elucidated.
Collapse
Affiliation(s)
| | - John James Zaunders
- Kirby Institute, University of New South Wales, 2031 Sydney, Australia.
- Centre for Applied Medical Research, St. Vincent's Hospital, 2010 Sydney, Australia.
| | - Anthony Dominic Kelleher
- Kirby Institute, University of New South Wales, 2031 Sydney, Australia.
- Centre for Applied Medical Research, St. Vincent's Hospital, 2010 Sydney, Australia.
| |
Collapse
|
18
|
Lokhande MU, Thimme R, Klenerman P, Semmo N. Methodologies for the Analysis of HCV-Specific CD4(+) T Cells. Front Immunol 2015; 6:57. [PMID: 25767470 PMCID: PMC4341113 DOI: 10.3389/fimmu.2015.00057] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 01/29/2015] [Indexed: 12/16/2022] Open
Abstract
Virus-specific CD4+ T cells play a major role in viral infections, such as hepatitis C virus (HCV). Viral clearance is associated with vigorous and multi-specific CD4+ T-cell responses, while chronic infection has been shown to be associated with weak or absent T-cell responses. Most of these studies have used functional assays to analyze virus-specific CD4+ T-cell responses; however, these and other detection methods have various limitations. Therefore, the important question of whether virus-specific CD4+ T cells are completely absent or primarily impaired in specific effector functions during chronic infection, has yet to be analyzed in detail. A novel assay, in which virus-specific CD4+ T-cell frequencies can be determined by de novo CD154 (CD40 ligand) expression in response to viral antigens, can help to overcome some of the limitations of functional assays and restrictions of multimer-based methods. This and other current established methods for the detection of HCV-specific CD4+ T cells will be discussed in this review.
Collapse
Affiliation(s)
- Megha U Lokhande
- Hepatology, Department of Clinical Research, University of Bern , Bern , Switzerland
| | - Robert Thimme
- Department of Gastroenterology and Hepatology, University Hospital of Freiburg , Freiburg , Germany
| | - Paul Klenerman
- NIHR Biomedical Research Centre, Oxford and Peter Medawar Building for Pathogen Research, University of Oxford , Oxford , UK
| | - Nasser Semmo
- Hepatology, Department of Clinical Research, University of Bern , Bern , Switzerland ; Department of Hepatology, University Clinic of Visceral Surgery and Medicine , Inselspital, Bern , Switzerland
| |
Collapse
|
19
|
Sugden PB, Cameron B, Mina M, Lloyd AR. Protection against hepatitis C infection via NK cells in highly-exposed uninfected injecting drug users. J Hepatol 2014; 61:738-45. [PMID: 24845613 DOI: 10.1016/j.jhep.2014.05.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 04/17/2014] [Accepted: 05/06/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS HCV seroprevalence surveys in longstanding injecting drug users (IDUs) reveal a small minority who remain seronegative, with some exhibiting HCV-specific cellular immunity. This study aimed to characterise this immunity, assess associations with risk behaviours and protection against infection. METHODS A nested case-control series from a prospective cohort of seronegative IDUs was selected with incident cases (IN; n = 28) matched by demographics and risk behaviour to exposed uninfected (EU) subjects (n = 28). Samples were assayed for natural killer (NK) cell phenotypes and function, HCV-specific IFNγ in ELISpot, and HCV-specific CD4 T effector responses. IL28B and HLA-C/KIR2DL3 genotypes were tested. RESULTS Numbers of activated (CD69(+)) NK cells in the mature CD56(dim)CD16(+) subset, and cytotoxic (NKp30(+)) cells in the CD56(bright)CD16(+) subset were higher in the EU subjects (p = 0.040, p = 0.038 respectively). EU subjects had higher frequencies of interferon gamma (IFNγ) producing NK cells, and lower frequencies of CD107a expression (p = 0.003, p = 0.015 respectively). By contrast, the frequency, magnitude, and breadth of HCV-specific CD4 and CD8 T cell responses did not differ, nor did IL28B, HLA-C, or KIR2DL3 allele frequencies. CONCLUSIONS Sustained NK cell activation contributes to protection against HCV infection. HCV-specific cellular immunity is prevalent in EU subjects but does not appear to be protective.
Collapse
MESH Headings
- Adult
- Antigens, CD/immunology
- Antigens, Differentiation, T-Lymphocyte/immunology
- Drug Users/psychology
- Female
- Gene Expression Profiling
- Hepatitis C/etiology
- Hepatitis C/genetics
- Hepatitis C/immunology
- Hepatitis C/prevention & control
- Humans
- Interferons
- Interleukins/genetics
- Interleukins/immunology
- Killer Cells, Natural/immunology
- Lectins, C-Type/immunology
- Lymphocyte Activation/immunology
- Male
- Natural Cytotoxicity Triggering Receptor 3/immunology
- Receptors, KIR2DL3/genetics
- Receptors, KIR2DL3/immunology
- Risk-Taking
- Substance Abuse, Intravenous/complications
- Substance Abuse, Intravenous/genetics
- Substance Abuse, Intravenous/immunology
- Substance Abuse, Intravenous/psychology
Collapse
Affiliation(s)
- Peter B Sugden
- Inflammation and Infection Research Centre, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Barbara Cameron
- Inflammation and Infection Research Centre, School of Medical Sciences, University of New South Wales, Sydney, Australia.
| | - Michael Mina
- Inflammation and Infection Research Centre, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Andrew R Lloyd
- Inflammation and Infection Research Centre, School of Medical Sciences, University of New South Wales, Sydney, Australia
| |
Collapse
|
20
|
Tong WWY, Shepherd K, Garland S, Meagher A, Templeton DJ, Fairley CK, Jin F, Poynten IM, Zaunders J, Hillman RJ, Grulich AE, Kelleher AD, Carr A. Human Papillomavirus 16–Specific T-Cell Responses and Spontaneous Regression of Anal High-Grade Squamous Intraepithelial Lesions. J Infect Dis 2014; 211:405-15. [DOI: 10.1093/infdis/jiu461] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
21
|
Sadler R, Bateman EAL, Heath V, Patel SY, Schwingshackl PP, Cullinane AC, Ayers L, Ferry BL. Establishment of a healthy human range for the whole blood "OX40" assay for the detection of antigen-specific CD4+ T cells by flow cytometry. CYTOMETRY PART B-CLINICAL CYTOMETRY 2014; 86:350-61. [PMID: 24827553 DOI: 10.1002/cyto.b.21165] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 12/17/2013] [Accepted: 01/29/2014] [Indexed: 12/21/2022]
Abstract
BACKGROUND Clinical investigation of antigen-specific T cells in potentially immunodeficient patients is an important and often challenging aspect of patient diagnostic work up. Methods for detection of microbial exposure to the T-cell compartment exist but are laborious and time consuming. Recently, a whole blood technique involving flow cytometry and detection of CD25 and OX40 (CD134) expression on the surface of activated CD4+ T cells was shown to be accurate and concordant when compared with more traditional methods of antigen-specific T-cell detection. METHODS Whole heparinized blood was collected from healthy donors and set up using the "OX40" assay to detect antigen-specific CD4+ T-cell responses to Varicella Zoster Virus, Epstein-Barr Virus (EBV), Cytomegalovirus, Candida albicans, and Streptococcus pneumoniae. RESULTS The "OX40" assay technique was clinically validated for routine use in an NHS clinical immunology laboratory by analysis of incubation length (40-50 h), sample transport time (up to 24 h at room temperature), concordance with serology testing, proliferation and interferon-gamma production. In addition, 63 healthy controls (age range 21-78) were tested for responses to generate a healthy control reference range. CONCLUSIONS The OX40 assay, as presented in this report, represents an economical, rapid, robust whole blood technique to detect antigen-specific T cells, which is suitable for clinical immunology diagnostic laboratory use.
Collapse
Affiliation(s)
- Ross Sadler
- Department of Immunology, Churchill Hospital, Oxford University Hospitals NHS Trust, Oxford, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Seddiki N, Cook L, Hsu DC, Phetsouphanh C, Brown K, Xu Y, Kerr SJ, Cooper DA, Munier CML, Pett S, Ananworanich J, Zaunders J, Kelleher AD. Human antigen-specific CD4+CD25+CD134+CD39+T cells are enriched for regulatory T cells and comprise a substantial proportion of recall responses. Eur J Immunol 2014; 44:1644-61. [DOI: 10.1002/eji.201344102] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 01/28/2014] [Accepted: 03/12/2014] [Indexed: 02/04/2023]
Affiliation(s)
- Nabila Seddiki
- The Kirby Institute; University of New South Wales; Sydney NSW Australia
- St Vincent's Centre for Applied Medical Research; Sydney NSW Australia
| | - Laura Cook
- The Kirby Institute; University of New South Wales; Sydney NSW Australia
- St Vincent's Centre for Applied Medical Research; Sydney NSW Australia
| | - Denise C. Hsu
- The Kirby Institute; University of New South Wales; Sydney NSW Australia
- St Vincent's Centre for Applied Medical Research; Sydney NSW Australia
- HIV Netherlands Australia Thailand Research Collaboration; Thai Red Cross AIDS Research Centre; Bangkok Thailand
| | - Chansavath Phetsouphanh
- The Kirby Institute; University of New South Wales; Sydney NSW Australia
- St Vincent's Centre for Applied Medical Research; Sydney NSW Australia
| | - Kai Brown
- The Kirby Institute; University of New South Wales; Sydney NSW Australia
- St Vincent's Centre for Applied Medical Research; Sydney NSW Australia
| | - Yin Xu
- The Kirby Institute; University of New South Wales; Sydney NSW Australia
- St Vincent's Centre for Applied Medical Research; Sydney NSW Australia
| | - Stephen J. Kerr
- The Kirby Institute; University of New South Wales; Sydney NSW Australia
- St Vincent's Centre for Applied Medical Research; Sydney NSW Australia
- HIV Netherlands Australia Thailand Research Collaboration; Thai Red Cross AIDS Research Centre; Bangkok Thailand
| | - David A. Cooper
- The Kirby Institute; University of New South Wales; Sydney NSW Australia
- St Vincent's Centre for Applied Medical Research; Sydney NSW Australia
- HIV; Immunology and Infectious Diseases Clinical Services Unit; St Vincent's Hospital; Sydney NSW Australia
| | - C. Mee Ling Munier
- The Kirby Institute; University of New South Wales; Sydney NSW Australia
| | - Sarah Pett
- St Vincent's Centre for Applied Medical Research; Sydney NSW Australia
- HIV; Immunology and Infectious Diseases Clinical Services Unit; St Vincent's Hospital; Sydney NSW Australia
| | - Jintanat Ananworanich
- HIV Netherlands Australia Thailand Research Collaboration; Thai Red Cross AIDS Research Centre; Bangkok Thailand
| | - John Zaunders
- The Kirby Institute; University of New South Wales; Sydney NSW Australia
| | - Anthony D. Kelleher
- The Kirby Institute; University of New South Wales; Sydney NSW Australia
- St Vincent's Centre for Applied Medical Research; Sydney NSW Australia
- HIV; Immunology and Infectious Diseases Clinical Services Unit; St Vincent's Hospital; Sydney NSW Australia
| |
Collapse
|
23
|
Hsu DC, Kerr SJ, Thongpaeng P, Iampornsin T, Pett SL, Zaunders JJ, Avihingsanon A, Ubolyam S, Ananworanich J, Kelleher AD, Cooper DA. Incomplete restoration of Mycobacterium tuberculosis-specific-CD4 T cell responses despite antiretroviral therapy. J Infect 2013; 68:344-54. [PMID: 24325926 DOI: 10.1016/j.jinf.2013.11.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 11/26/2013] [Accepted: 11/29/2013] [Indexed: 11/24/2022]
Abstract
OBJECTIVES Despite antiretroviral therapy (ART), HIV-infected persons have increased risk of active tuberculosis (TB). PPD and combined ESAT-6 and CFP-10-specific-CD4 (EC-Sp-CD4) responses were examined over 96 weeks. METHODS HIV-infected, ART-naive Thai adults with CD4 T cell count ≤350 cells/μL starting ART were assessed at baseline, wk4, 8, 12, 24, 48 and 96. PPD and EC-Sp-CD4 T cells were detected by CD25/CD134 co-expression after stimulation with antigens. RESULTS Fifty subjects were enrolled, 39 were male, median age 32 yrs, median baseline CD4 T cell count 186 cells/μL and plasma HIV-viral-load 4.9log10 copies/mL. Seventeen were TB-sensitised. At baseline, 25 had positive PPD and 15 had positive EC-Sp-CD4 response. CD4 T cell count <100 cells/μL was less (P = 0.005) and TB-sensitisation was more likely (P = 0.013) to be associated with positive baseline PPD-Sp-CD4 response. At wk4, the number of subjects with positive PPD-Sp-CD4 response rose to 35 (P = 0.021). Mean PPD-Sp-CD4 T cells increased at wk4 (P = 0.017) in patients not classified as TB-sensitised. The number of subjects with positive EC-Sp-CD4 response did not change significantly post ART. In TB-sensitised patients, mean EC-Sp-CD4 T cells declined to below baseline from wk12 (P = 0.010) onwards. EC-Sp-CD4 responses were undetectable in 3 out of 17 TB-sensitised patients. CONCLUSIONS Restoration of responses to TB-antigens was incomplete and inconsistent under the employed experimental conditions and may account for persistent increased risk of TB despite ART.
Collapse
Affiliation(s)
- Denise C Hsu
- The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, Australia; HIV Netherlands Australia Thailand Research Collaboration, Thai Red Cross AIDS Research Centre, Bangkok, Thailand.
| | - Stephen J Kerr
- The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, Australia; HIV Netherlands Australia Thailand Research Collaboration, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Parawee Thongpaeng
- HIV Netherlands Australia Thailand Research Collaboration, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Thatri Iampornsin
- HIV Netherlands Australia Thailand Research Collaboration, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Sarah L Pett
- The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, Australia; St Vincent's Centre for Applied Medical Research, Sydney, Australia; UCL Research Department of Infection and Population Health, UCL, London, United Kingdom
| | - John J Zaunders
- The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, Australia; St Vincent's Centre for Applied Medical Research, Sydney, Australia
| | - Anchalee Avihingsanon
- HIV Netherlands Australia Thailand Research Collaboration, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Sasiwimol Ubolyam
- HIV Netherlands Australia Thailand Research Collaboration, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Jintanat Ananworanich
- HIV Netherlands Australia Thailand Research Collaboration, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Anthony D Kelleher
- The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, Australia; St Vincent's Centre for Applied Medical Research, Sydney, Australia
| | - David A Cooper
- The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, Australia; St Vincent's Centre for Applied Medical Research, Sydney, Australia
| |
Collapse
|
24
|
Hsu DC, Kerr SJ, Iampornsin T, Pett SL, Avihingsanon A, Thongpaeng P, Zaunders JJ, Ubolyam S, Ananworanich J, Kelleher AD, Cooper DA. Restoration of CMV-specific-CD4 T cells with ART occurs early and is greater in those with more advanced immunodeficiency. PLoS One 2013; 8:e77479. [PMID: 24130889 PMCID: PMC3795037 DOI: 10.1371/journal.pone.0077479] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 09/02/2013] [Indexed: 12/21/2022] Open
Abstract
Objectives Restoration of Cytomegalovirus-specific-CD4 T cell (CMV-Sp-CD4) responses partly accounts for the reduction of CMV-disease with antiretroviral-therapy (ART), but CMV-Sp-CD4 may also drive immune activation and immunosenescence. This study characterized the dynamics of CMV-Sp-CD4 after ART initiation and explored associations with CD4 T cell recovery as well as frequency of naïve CD4 T cells at week 96. Methods Fifty HIV-infected, ART-naïve Thai adults with CD4 T cell count ≤350cells/µL and starting ART were evaluated over 96 weeks (ClinicalTrials.gov identifier NCT01296373). CMV-Sp-CD4 was detected by co-expression of CD25/CD134 by flow cytometry after CMV-antigen stimulation. Results All subjects were CMV sero-positive, 4 had quantifiable CMV-DNA (range 2.3-3.9 log10 copies/mL) at baseline but none had clinically apparent CMV-disease. Baseline CMV-Sp-CD4 response was positive in 40 subjects. Those with CD4 T cell count <100cells/µL were less likely to have positive baseline CMV-Sp-CD4 response (P=0.003). Positive baseline CMV-Sp-CD4 response was associated with reduced odds of quantifiable CMV-DNA (P=0.022). Mean CD4 T cell increase at week 96 was 213 cells/µL. This was associated positively with baseline HIV-VL (P=0.001) and negatively with age (P=0.003). The frequency of CMV-Sp-CD4 increased at week 4 (P=0.008), then declined. Those with lower baseline CMV-Sp-CD4 (P=0.009) or CDC category C (P<0.001) had greater increases in CMV-Sp-CD4 at week 4. At week 96, CD4 T cell count was positively (P<0.001) and the frequency of CMV-Sp-CD4 was negatively (P=0.001) associated with the percentage of naïve CD4 T cells. Conclusions Increases in CMV-Sp-CD4 with ART occurred early and were greater in those with more advanced immunodeficiency. The frequency of CMV-Sp-CD4 was associated with reduced naïve CD4 T cells, a marker associated with immunosenescence.
Collapse
Affiliation(s)
- Denise C. Hsu
- The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, Australia
- HIV Netherlands Australia Thailand Research Collaboration, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
- * E-mail:
| | - Stephen J. Kerr
- The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, Australia
- HIV Netherlands Australia Thailand Research Collaboration, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Thatri Iampornsin
- HIV Netherlands Australia Thailand Research Collaboration, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Sarah L. Pett
- The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, Australia
- St Vincent’s Centre for Applied Medical Research, Sydney, Australia
| | - Anchalee Avihingsanon
- HIV Netherlands Australia Thailand Research Collaboration, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Parawee Thongpaeng
- HIV Netherlands Australia Thailand Research Collaboration, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - John J. Zaunders
- The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, Australia
- St Vincent’s Centre for Applied Medical Research, Sydney, Australia
| | - Sasiwimol Ubolyam
- HIV Netherlands Australia Thailand Research Collaboration, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Jintanat Ananworanich
- HIV Netherlands Australia Thailand Research Collaboration, Thai Red Cross AIDS Research Centre, Bangkok, Thailand
| | - Anthony D. Kelleher
- The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, Australia
- St Vincent’s Centre for Applied Medical Research, Sydney, Australia
| | - David A. Cooper
- The Kirby Institute for Infection and Immunity in Society, University of New South Wales, Sydney, Australia
- St Vincent’s Centre for Applied Medical Research, Sydney, Australia
| |
Collapse
|
25
|
Phetsouphanh C, Xu Y, Amin J, Seddiki N, Procopio F, Sekaly RP, Zaunders JJ, Kelleher AD. Characterization of transcription factor phenotypes within antigen-specific CD4+ T cells using qualitative multiplex single-cell RT-PCR. PLoS One 2013; 8:e74946. [PMID: 24124462 PMCID: PMC3790772 DOI: 10.1371/journal.pone.0074946] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2013] [Accepted: 08/07/2013] [Indexed: 11/25/2022] Open
Abstract
Current research on antigen specific CD4+ T cells indicates that there is functional and phenotypic heterogeneity within these populations, but the extent of this heterogeneity is poorly described. The CD134/CD25 assay allows live isolation of antigen specific cells in vitro for down-stream molecular analysis. Antigen specific CD4+ T cells were examined at the molecular level by lineage specific transcription factor profiling using qualitative multiplex single cell RT-PCR and Lock Nucleic Acid (LNA) probes allowed unbiased amplification and delineation of expression of Tbx21, Gata3, Rorc, Foxp3 and Bcl-6. It overcomes the limitations of previous assays by allowing identification of transcription factor mRNA in single antigen specific cells with high sensitivity (down to 10 femtograms) and specificity. Patterns of responses can be robustly characterized using <200 cells based on exact binomial calculations. These results are reproducible with a CV of ≈6%. The patterns of heterogeneity are stable within an individual antigen specific response but vary between responses to different antigens. Responses to CMV have a Th1 predominant profile (35.6% of responding cells expressing tbx21) whereas responses to Tetanus Toxoid have a Th2 biased profile (22% of responding cells expressing gata3), with unexpectedly high levels of Treg cells found in both populations. Here we describe a methodology that allows live isolation of Ag specific cells and transcription factor profiling at a single cell level to robustly delineate the different CD4+ T cell subsets within this population. This novel method is a powerful tool that can be used to study CD4+ T cell heterogeneity within extremely small populations of cells and where cell numbers are limited.
Collapse
Affiliation(s)
- Chansavath Phetsouphanh
- Kirby Institute, University of New South Wales, Sydney, Australia
- Centre for Applied Medical Research, St Vincent's Hospital, Sydney, Australia
- * E-mail:
| | - Yin Xu
- Kirby Institute, University of New South Wales, Sydney, Australia
- Centre for Applied Medical Research, St Vincent's Hospital, Sydney, Australia
| | - Janaki Amin
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Nabila Seddiki
- Kirby Institute, University of New South Wales, Sydney, Australia
- Centre for Applied Medical Research, St Vincent's Hospital, Sydney, Australia
| | - Francesco Procopio
- Vaccine and Gene Therapy Institute (VGTI), Port St. Lucie, Florida, United States of America
| | - Rafick Pierre Sekaly
- Vaccine and Gene Therapy Institute (VGTI), Port St. Lucie, Florida, United States of America
| | - John J. Zaunders
- Kirby Institute, University of New South Wales, Sydney, Australia
- Centre for Applied Medical Research, St Vincent's Hospital, Sydney, Australia
| | - Anthony D. Kelleher
- Kirby Institute, University of New South Wales, Sydney, Australia
- Centre for Applied Medical Research, St Vincent's Hospital, Sydney, Australia
| |
Collapse
|
26
|
Chang CC, Crane M, Zhou J, Mina M, Post JJ, Cameron BA, Lloyd AR, Jaworowski A, French MA, Lewin SR. HIV and co-infections. Immunol Rev 2013; 254:114-42. [PMID: 23772618 PMCID: PMC3697435 DOI: 10.1111/imr.12063] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite significant reductions in morbidity and mortality secondary to availability of effective combination anti-retroviral therapy (cART), human immunodeficiency virus (HIV) infection still accounts for 1.5 million deaths annually. The majority of deaths occur in sub-Saharan Africa where rates of opportunistic co-infections are disproportionately high. In this review, we discuss the immunopathogenesis of five common infections that cause significant morbidity in HIV-infected patients globally. These include co-infection with Mycobacterium tuberculosis, Cryptococcus neoformans, hepatitis B virus, hepatitis C virus, and Plasmodium falciparum. Specifically, we review the natural history of each co-infection in the setting of HIV, the specific immune defects induced by HIV, the effects of cART on the immune response to the co-infection, the pathogenesis of immune restoration disease (IRD) associated with each infection, and advances in the areas of prevention of each co-infection via vaccination. Finally, we discuss the opportunities and gaps in knowledge for future research.
Collapse
Affiliation(s)
- Christina C Chang
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Chang CC, Lim A, Omarjee S, Levitz SM, Gosnell BI, Spelman T, Elliott JH, Carr WH, Moosa MYS, Ndung'u T, Lewin SR, French MA. Cryptococcosis-IRIS is associated with lower cryptococcus-specific IFN-γ responses before antiretroviral therapy but not higher T-cell responses during therapy. J Infect Dis 2013; 208:898-906. [PMID: 23766525 DOI: 10.1093/infdis/jit271] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Cryptococcosis-associated immune reconstitution inflammatory syndrome (C-IRIS) may be driven by aberrant T-cell responses against cryptococci. We investigated this in human immunodeficiency virus (HIV)-infected patients with treated cryptococcal meningitis (CM) commencing combination antiretroviral therapy (cART). METHODS Mitogen- and cryptococcal mannoprotein (CMP)-activated (CD25+CD134+) CD4+ T cells and -induced production of interferon-gamma (IFN-γ), IL-10, and CXCL10 were assessed in whole blood cultures in a prospective study of 106 HIV-CM coinfected patients. RESULTS Patients with paradoxical C-IRIS (n = 27), compared with patients with no neurological deterioration (no ND; n = 63), had lower CMP-induced IFN-γ production in 24-hour cultures pre-cART and 4 weeks post-cART (P = .0437 and .0257, respectively) and lower CMP-activated CD4+ T-cell counts pre-cART (P = .0178). Patients surviving to 24 weeks had higher proportions of mitogen-activated CD4+ T cells and higher CMP-induced CXCL10 and IL-10 production in 24-hour cultures pre-cART than patients not surviving (P = .0053, .0436 and .0319, respectively). C-IRIS was not associated with higher CMP-specific T-cell responses before or during cART. CONCLUSION Greater preservation of T-cell function and higher CMP-induced IL-10 and CXCL10 production before cART are associated with improved survival while on cART. Lower CMP-induced IFN-γ production pre-cART, but not higher CMP-specific T-cell responses after cART, were risk factors for C-IRIS.
Collapse
Affiliation(s)
- Christina C Chang
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|