1
|
Kälble F, Leonhard J, Zeier M, Zivanovic O, Schaier M, Steinborn A. Exhaustion of CD8 pos central memory regulatory T cell differentiation is involved in renal allograft rejection. Front Immunol 2025; 16:1532086. [PMID: 39925813 PMCID: PMC11802571 DOI: 10.3389/fimmu.2025.1532086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/03/2025] [Indexed: 02/11/2025] Open
Abstract
Background The role of regulatory CD8pos T cells (CD8pos Tregs) and cytotoxic CD8pos responder T cells (CD8pos Tresps) in maintaining stable graft function in kidney transplant recipients (KTR) remains largely unclear. The pathogenesis of graft deterioration in case of rejection involves the exhaustive differentiation of both CD8pos T cell subsets, but the causal mechanisms have not yet been identified. Methods In this study, we separately investigated the differentiation of CD8posTregs/Tresps in 134 stable KTR with no evidence of renal graft rejection, in 41 KTR diagnosed with biopsy-confirmed rejection at enrolment and in 5 patients who were unremarkable at enrolment, but developed rejection within three years of enrolment. We were investigating whether changed differentiation of CCR7posCD45RAposCD31pos recent thymic emigrant (RTE) cells via CD45RAnegCD31pos memory (CD31pos memory) cells (pathway 1), via direct proliferation (pathway 2), or via CCR7posCD45RA+CD31neg resting mature naïve (MN) cells (pathway 3) into CD45RAnegCD31neg memory (CD31neg memory) cells affects the CD8pos Treg/Tresp ratio or identifies a CD8pos Treg/Tresp subset that predicts or confirms renal allograft rejection. Results We found that RTE Treg differentiation via pathway 1 was age-independently increased in KTR, who developed graft rejection during the follow-up period, leading to abundant MN Treg and central memory Treg (CM Treg) production and favoring a strongly increased CD8pos Treg/Tresp ratio. In KTR with biopsy-confirmed rejection at the time of enrolment, an increased differentiation of RTE Tregs into CCR7negCD45RAposCD31neg terminally differentiated effector memory (CD31neg TEMRA Tregs) and CD31pos memory Tregs was observed. CD31neg memory Treg production was maintained by alternative differentiation of resting MN Tregs, resulting in increased effector memory Treg (EM Treg) production, while the CD8pos Treg/Treg ratio was unaffected. An altered differentiation of CD8pos Tresps was not observed, shifting the Treg/Tresp ratio in favor of Tregs. Conclusions Our results show that exhaustive CD8pos Treg differentiation into CM Tregs may lead to future rejection, with a shift towards EM Treg production and an accumulation of CD31neg TEMRA Tregs in KTR with current rejection.
Collapse
Affiliation(s)
- Florian Kälble
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - Jonas Leonhard
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Martin Zeier
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - Oliver Zivanovic
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| | - Matthias Schaier
- Department of Nephrology, University of Heidelberg, Heidelberg, Germany
| | - Andrea Steinborn
- Department of Obstetrics and Gynecology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
2
|
Larbi A. From Genesis to Old Age: Exploring the Immune System One Cell at a Time with Flow Cytometry. Biomedicines 2024; 12:1469. [PMID: 39062042 PMCID: PMC11275137 DOI: 10.3390/biomedicines12071469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/21/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
The immune system is a highly complex and tightly regulated system that plays a crucial role in protecting the body against external threats, such as pathogens, and internal abnormalities, like cancer cells. It undergoes development during fetal stages and continuously learns from each encounter with pathogens, allowing it to develop immunological memory and provide a wide range of immune protection. Over time, after numerous encounters and years of functioning, the immune system can begin to show signs of erosion, which is commonly named immunosenescence. In this review, we aim to explore how the immune system responds to initial encounters with antigens and how it handles persistent stimulations throughout a person's lifetime. Our understanding of the immune system has greatly benefited from advanced technologies like flow cytometry. In this context, we will discuss the valuable contribution of flow cytometry in enhancing our knowledge of the immune system behavior in aging, with a specific focus on T-cells. Moreover, we will expand our discussion to the flow cytometry-based assessment of extracellular vesicles, a recently discovered communication channel in biology, and their implications for immune system functioning.
Collapse
Affiliation(s)
- Anis Larbi
- Medical and Scientific Affairs, Beckman Coulter Life Sciences, 22 Avenue des Nations, 93420 Villepinte, France;
- Department of Medicine, Division of Geriatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| |
Collapse
|
3
|
Boßlau TK, Wasserfurth P, Reichel T, Weyh C, Palmowski J, Nebl J, Joisten N, Belen S, Schenk A, Hahn A, Zimmer P, Krüger K. 12-week combined strength and endurance exercise attenuates CD8 + T-cell differentiation and affects the kynurenine pathway in the elderly: a randomized controlled trial. Immun Ageing 2023; 20:19. [PMID: 37161540 PMCID: PMC10169370 DOI: 10.1186/s12979-023-00347-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 05/03/2023] [Indexed: 05/11/2023]
Abstract
BACKGROUND Age-related accumulation of highly differentiated CD8+ effector memory re-expressing CD45RA (EMRA) T-cells and disruption of the kynurenine (KYN) pathway are associated with chronic inflammation and the development of insulin resistance. In this study the aim was to investigate the effects of 12-week combined strength and endurance exercise on CD8+ T-cell differentiation and KYN pathway metabolites. Ninety-six elderly subjects (f/m, aged 50-70) were randomized to a control (CON) or exercise (EX) group. The EX group completed combined strength and endurance training twice weekly for one hour each time at an intensity of 60% of the one-repetition maximum for strength exercises and a perceived exertion of 15/20 for endurance exercises. The EX group was also randomly subdivided into two groups with or without a concomitant balanced diet intervention in order to examine additional effects besides exercise alone. Before and after the intervention phase, the proportions of CD8+ T-cell subsets and levels of KYN pathway metabolites in peripheral blood were determined. RESULTS The CD8+ EMRA T-cell subsets increased in the CON group but remained almost unchanged in the EX group (p = .02). Plasma levels of kynurenic acid (KA) increased in the EX group and decreased in the CON group (p = .03). Concomitant nutritional intervention resulted in lower levels of quinolinic acid (QA) compared with exercise alone (p = .03). Overall, there was a slight increase in the QA/KA ratio in the CON group, whereas it decreased in the EX group (p > .05). CONCLUSIONS Combined strength and endurance training seems to be a suitable approach to attenuate CD8+ T-cell differentiation in the elderly and to redirect the KYN pathway towards KA. The clinical relevance of these effects needs further investigation.
Collapse
Affiliation(s)
- Tim Konstantin Boßlau
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Kugelberg 62, 35394, Giessen, Germany.
| | - Paulina Wasserfurth
- Department of Exercise, Nutrition and Health, Faculty of Sport and Health Sciences, Technical University Munich, Connollystraße 32, 80809, Munich, Germany
| | - Thomas Reichel
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Kugelberg 62, 35394, Giessen, Germany
| | - Christopher Weyh
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Kugelberg 62, 35394, Giessen, Germany
| | - Jana Palmowski
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Kugelberg 62, 35394, Giessen, Germany
| | - Josefine Nebl
- Faculty of Natural Sciences, Institute of Food Science and Human Nutrition, Leibniz University Hanover, Am Kleinen Felde 30, 30159, Hannover, Germany
| | - Niklas Joisten
- Division of Performance and Health, Institute for Sport and Sport Science, Technical University Dortmund, Otto-Hahn-Str. 3, 44227, Dortmund, Germany
| | - Sergen Belen
- Division of Performance and Health, Institute for Sport and Sport Science, Technical University Dortmund, Otto-Hahn-Str. 3, 44227, Dortmund, Germany
| | - Alexander Schenk
- Division of Performance and Health, Institute for Sport and Sport Science, Technical University Dortmund, Otto-Hahn-Str. 3, 44227, Dortmund, Germany
| | - Andreas Hahn
- Faculty of Natural Sciences, Institute of Food Science and Human Nutrition, Leibniz University Hanover, Am Kleinen Felde 30, 30159, Hannover, Germany
| | - Philipp Zimmer
- Division of Performance and Health, Institute for Sport and Sport Science, Technical University Dortmund, Otto-Hahn-Str. 3, 44227, Dortmund, Germany
| | - Karsten Krüger
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Kugelberg 62, 35394, Giessen, Germany
| |
Collapse
|
4
|
Mestiri S, Merhi M, Inchakalody VP, Taib N, Smatti MK, Ahmad F, Raza A, Ali FH, Hydrose S, Fernandes Q, Ansari AW, Sahir F, Al-Zaidan L, Jalis M, Ghoul M, Allahverdi N, Al Homsi MU, Uddin S, Jeremijenko AM, Nimir M, Abu-Raddad LJ, Abid FB, Zaqout A, Alfheid SR, Saqr HMH, Omrani AS, Hssain AA, Al Maslamani M, Yassine HM, Dermime S. Persistence of spike-specific immune responses in BNT162b2-vaccinated donors and generation of rapid ex-vivo T cells expansion protocol for adoptive immunotherapy: A pilot study. Front Immunol 2023; 14:1061255. [PMID: 36817441 PMCID: PMC9933868 DOI: 10.3389/fimmu.2023.1061255] [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: 10/04/2022] [Accepted: 01/11/2023] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION The BNT162b2 mRNA-based vaccine has shown high efficacy in preventing COVID-19 infection but there are limited data on the types and persistence of the humoral and T cell responses to such a vaccine. METHODS Here, we dissect the vaccine-induced humoral and cellular responses in a cohort of six healthy recipients of two doses of this vaccine. RESULTS AND DISCUSSION Overall, there was heterogeneity in the spike-specific humoral and cellular responses among vaccinated individuals. Interestingly, we demonstrated that anti-spike antibody levels detected by a novel simple automated assay (Jess) were strongly correlated (r=0.863, P<0.0001) with neutralizing activity; thus, providing a potential surrogate for neutralizing cell-based assays. The spike-specific T cell response was measured with a newly modified T-spot assay in which the high-homology peptide-sequences cross-reactive with other coronaviruses were removed. This response was induced in 4/6 participants after the first dose, and all six participants after the second dose, and remained detectable in 4/6 participants five months post-vaccination. We have also shown for the first time, that BNT162b2 vaccine enhanced T cell responses also against known human common viruses. In addition, we demonstrated the efficacy of a rapid ex-vivo T cell expansion protocol for spike-specific T cell expansion to be potentially used for adoptive-cell therapy in severe COVID-19, immunocompromised individuals, and other high-risk groups. There was a 9 to 13.7-fold increase in the number of expanded T cells with a significant increase of anti-spike specific response showing higher frequencies of both activation and cytotoxic markers. Interestingly, effector memory T cells were dominant in all four participants' CD8+ expanded memory T cells; CD4+ T cells were dominated by effector memory in 2/4 participants and by central memory in the remaining two participants. Moreover, we found that high frequencies of CD4+ terminally differentiated memory T cells were associated with a greater reduction of spike-specific activated CD4+ T cells. Finally, we showed that participants who had a CD4+ central memory T cell dominance expressed a high CD69 activation marker in the CD4+ activated T cells.
Collapse
Affiliation(s)
- Sarra Mestiri
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Maysaloun Merhi
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Varghese P. Inchakalody
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Nassiba Taib
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Maria K. Smatti
- Qatar University Biomedical Research Center, Qatar University, Doha, Qatar
| | - Fareed Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Afsheen Raza
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Fatma H. Ali
- Qatar University Biomedical Research Center, Qatar University, Doha, Qatar
| | - Shereena Hydrose
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Queenie Fernandes
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- College of Medicine, Qatar University, Doha, Qatar
| | - Abdul W. Ansari
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Fairooz Sahir
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Lobna Al-Zaidan
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Munir Jalis
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Mokhtar Ghoul
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Niloofar Allahverdi
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Mohammed U. Al Homsi
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
- Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | | | - Mai Nimir
- Communicable Disease Center, Hamad Medical Corporation, Doha, Qatar
| | - Laith J. Abu-Raddad
- Infectious Disease Epidemiology Group, Weill Cornell Medicine–Qatar, Cornell University, Qatar Foundation–Education City, Doha, Qatar
- World Health Organization Collaborating Centre for Disease Epidemiology Analytics on HIV/AIDS, Sexually Transmitted Infections, and Viral Hepatitis, Weill Cornell Medicine–Qatar, Cornell University, Qatar Foundation–Education City, Doha, Qatar
- Department of Population Health Sciences, Weill Cornell Medicine, Cornell University, New York, NY, United States
| | - Fatma Ben Abid
- Communicable Disease Center, Hamad Medical Corporation, Doha, Qatar
| | - Ahmed Zaqout
- Communicable Disease Center, Hamad Medical Corporation, Doha, Qatar
| | | | | | - Ali S. Omrani
- College of Medicine, Qatar University, Doha, Qatar
- Communicable Disease Center, Hamad Medical Corporation, Doha, Qatar
| | - Ali Ait Hssain
- Medical Intensive Care Unit, Hamad Medical Corporation, Doha, Qatar
| | | | - Hadi M. Yassine
- Qatar University Biomedical Research Center, Qatar University, Doha, Qatar
| | - Said Dermime
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
5
|
Naik S, Triplett BM. Selective depletion of naïve T cells by targeting CD45RA. Front Oncol 2023; 12:1009143. [PMID: 36776371 PMCID: PMC9911795 DOI: 10.3389/fonc.2022.1009143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/22/2022] [Indexed: 01/28/2023] Open
Affiliation(s)
- Swati Naik
- *Correspondence: Swati Naik, ; Brandon M. Triplett,
| | | |
Collapse
|
6
|
Anmol K, Akanksha H, Zhengguo X. Are CD45RO+ and CD45RA- genuine markers for bovine memory T cells? ANIMAL DISEASES 2022. [DOI: 10.1186/s44149-022-00057-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
AbstractEffective vaccination induces memory T cells, which protect the host against pathogen re-infections. Therefore, detection of memory T cells is essential for evaluating vaccine efficacy, which was originally dependent on cytokine induction assays. Currently, two isoforms of CD45 tyrosine phosphatase, CD45RO expression and CD45RA exclusion (CD45RO+/ CD45RA-) are used extensively for detecting memory T cells in cattle. The CD45RO+/CD45RA- markers were first established in humans around three decades ago, and were adopted in cattle soon after. However, in the last two decades, some published data in humans have challenged the initial paradigm, and required multiple markers for identifying memory T cells. On the contrary, memory T cell detection in cattle still mostly relies on CD45RO+/CD45RA- despite some controversial evidence. In this review, we summarized the current literature to examine if CD45RO+/CD45RA- are valid markers for detecting memory T cells in cattle. It seems CD45RA and CD45RO (CD45RA/RO) as markers for identifying bovine memory T cells are questionable.
Collapse
|
7
|
Differential Expression of CD45RO and CD45RA in Bovine T Cells. Cells 2022; 11:cells11111844. [PMID: 35681539 PMCID: PMC9180881 DOI: 10.3390/cells11111844] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/28/2022] [Accepted: 06/02/2022] [Indexed: 01/06/2023] Open
Abstract
Effective vaccination induces immune memory to protect animals upon pathogen re-encounter. Despite contradictory reports, bovine memory T cells are identified based on two isoforms of CD45, expression of CD45RO plus exclusion of CD45RA. In this report, we contrasted CD45RA/RO expression on circulatory T cells with IFNγ and IL4 expression induced by a conventional method. To our surprise, 20% of cattle from an enclosed herd did not express CD45RO on T cells without any significant difference on CD45RA expression and IFNγ or IL4 induction. In CD45RO expressing cattle, CD45RA and CD45RO expressions excluded each other, with dominant CD45RO (>90%) expression on gamma delta (γδ) followed by CD4+ (60%) but significantly higher CD45RA expression on CD8+ T cells (about 80%). Importantly, more than 80% of CD45RO expressing CD4+ and CD8+ T cells failed to produce IFNγ and IL-4; however, within the cytokine inducing cells, CD4+ T cells highly expressed CD45RO but those within CD8+ T cells mostly expressed CD45RA. Hence, CD45RO is not ubiquitously expressed in cattle, and rather than with memory phenotype, CD45RA/RO expression are more associated with distinct T cell subtypes.
Collapse
|
8
|
Panoskaltsis N, McCarthy NE, Stagg AJ, Mummery CJ, Husni M, Arebi N, Greenstein D, Price CL, Al-Hassi HO, Koutinas M, Mantalaris A, Knight SC. Immune reconstitution and clinical recovery following anti-CD28 antibody (TGN1412)-induced cytokine storm. Cancer Immunol Immunother 2021; 70:1127-1142. [PMID: 33033851 PMCID: PMC7543968 DOI: 10.1007/s00262-020-02725-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 09/11/2020] [Indexed: 12/21/2022]
Abstract
Cytokine storm can result from cancer immunotherapy or certain infections, including COVID-19. Though short-term immune-related adverse events are routinely described, longer-term immune consequences and sequential immune monitoring are not as well defined. In 2006, six healthy volunteers received TGN1412, a CD28 superagonist antibody, in a first-in-man clinical trial and suffered from cytokine storm. After the initial cytokine release, antibody effect-specific immune monitoring started on Day + 10 and consisted mainly of evaluation of dendritic cell and T-cell subsets and 15 serum cytokines at 21 time-points over 2 years. All patients developed problems with concentration and memory; three patients were diagnosed with mild-to-moderate depression. Mild neutropenia and autoantibody production was observed intermittently. One patient suffered from peripheral dry gangrene, required amputations, and had persistent Raynaud's phenomenon. Gastrointestinal irritability was noted in three patients and coincided with elevated γδT-cells. One had pruritus associated with elevated IgE levels, also found in three other asymptomatic patients. Dendritic cells, initially undetectable, rose to normal within a month. Naïve CD8+ T-cells were maintained at high levels, whereas naïve CD4+ and memory CD4+ and CD8+ T-cells started high but declined over 2 years. T-regulatory cells cycled circannually and were normal in number. Cytokine dysregulation was especially noted in one patient with systemic symptoms. Over a 2-year follow-up, cognitive deficits were observed in all patients following TGN1412 infusion. Some also had signs or symptoms of psychological, mucosal or immune dysregulation. These observations may discern immunopathology, treatment targets, and long-term monitoring strategies for other patients undergoing immunotherapy or with cytokine storm.
Collapse
Affiliation(s)
- Nicki Panoskaltsis
- Department of Haematology, Imperial College London, Northwick Park & St. Mark's Campus, London, UK.
- Antigen Presentation Research Group, Imperial College London, Northwick Park & St. Mark's Campus, London, UK.
- Biological Systems Engineering Laboratory, Centre for Process Systems Engineering, Department of Chemical Engineering, Imperial College London, London, UK.
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, USA.
- BioMedical Systems Engineering Laboratory, Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, USA.
| | - Neil E McCarthy
- Antigen Presentation Research Group, Imperial College London, Northwick Park & St. Mark's Campus, London, UK
- Centre for Immunobiology, The Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Andrew J Stagg
- Antigen Presentation Research Group, Imperial College London, Northwick Park & St. Mark's Campus, London, UK
- Centre for Immunobiology, The Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Catherine J Mummery
- Dementia Research Centre, National Hospital for Neurology and Neurosurgery, Queen Square, Department of Neurology, Northwick Park Hospital, London, UK
- National Hospital for Neurology and Neurosurgery, University College London Hospital, University College London, London, UK
| | - Mariwan Husni
- Central and North West London Mental Health NHS Foundation Trust, Northwick Park Hospital, London, UK
- Psychiatry Department, Arabian Gulf University, Manama, Kingdom of Bahrain
| | - Naila Arebi
- Department of Gastroenterology and Intestinal Physiology, St. Mark's Hospital, London, UK
- Inflammatory Bowel Disease Clinical Service, St Mark's Hospital, London, UK
| | - David Greenstein
- Department of Vascular Surgery, North West London Hospitals NHS Trust, Northwick Park & St. Mark's Hospitals Site, London, UK
- Department of Vascular Surgery, Northwick Park Hospital and Imperial College London, London, UK
| | - Claire L Price
- Antigen Presentation Research Group, Imperial College London, Northwick Park & St. Mark's Campus, London, UK
- Lucid Group Communications, Buckinghamshire, UK
| | - Hafid O Al-Hassi
- Antigen Presentation Research Group, Imperial College London, Northwick Park & St. Mark's Campus, London, UK
- Research Institute in Healthcare Science, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, UK
| | - Michalis Koutinas
- Biological Systems Engineering Laboratory, Centre for Process Systems Engineering, Department of Chemical Engineering, Imperial College London, London, UK
- Department of Chemical Engineering, Cyprus University of Technology, Limassol, Cyprus
| | - Athanasios Mantalaris
- Biological Systems Engineering Laboratory, Centre for Process Systems Engineering, Department of Chemical Engineering, Imperial College London, London, UK
- BioMedical Systems Engineering Laboratory, Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, USA
| | - Stella C Knight
- Antigen Presentation Research Group, Imperial College London, Northwick Park & St. Mark's Campus, London, UK
| |
Collapse
|
9
|
Migueles SA, Rogan DC, Gavil NV, Kelly EP, Toulmin SA, Wang LT, Lack J, Ward AJ, Pryal PF, Ludwig AK, Medina RG, Apple BJ, Toumanios CN, Poole AL, Rehm CA, Jones SE, Liang CJ, Connors M. Antigenic Restimulation of Virus-Specific Memory CD8 + T Cells Requires Days of Lytic Protein Accumulation for Maximal Cytotoxic Capacity. J Virol 2020; 94:e01595-20. [PMID: 32907983 PMCID: PMC7654275 DOI: 10.1128/jvi.01595-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 09/05/2020] [Indexed: 02/07/2023] Open
Abstract
In various infections or vaccinations of mice or humans, reports of the persistence and the requirements for restimulation of the cytotoxic mediators granzyme B (GrB) and perforin (PRF) in CD8+ T cells have yielded disparate results. In this study, we examined the kinetics of PRF and GrB mRNA and protein expression after stimulation and associated changes in cytotoxic capacity in virus-specific memory cells in detail. In patients with controlled HIV or cleared respiratory syncytial virus (RSV) or influenza virus infections, all virus-specific CD8+ T cells expressed low PRF levels without restimulation. Following stimulation, they displayed similarly delayed kinetics for lytic protein expression, with significant increases occurring by days 1 to 3 before peaking on days 4 to 6. These increases were strongly correlated with, but were not dependent upon, proliferation. Incremental changes in PRF and GrB percent expression and mean fluorescence intensity (MFI) were highly correlated with increases in HIV-specific cytotoxicity. mRNA levels in HIV-specific CD8+ T-cells exhibited delayed kinetics after stimulation as with protein expression, peaking on day 5. In contrast to GrB, PRF mRNA transcripts were little changed over 5 days of stimulation (94-fold versus 2.8-fold, respectively), consistent with posttranscriptional regulation. Changes in expression of some microRNAs, including miR-17, miR-150, and miR-155, suggested that microRNAs might play a significant role in regulation of PRF expression. Therefore, under conditions of extremely low or absent antigen levels, memory virus-specific CD8+ T cells require prolonged stimulation over days to achieve maximal lytic protein expression and cytotoxic capacity.IMPORTANCE Antigen-specific CD8+ T cells play a major role in controlling most virus infections, primarily by perforin (PRF)- and granzyme B (GrB)-mediated apoptosis. There is considerable controversy regarding whether PRF is constitutively expressed, rapidly increased similarly to a cytokine, or delayed in its expression with more prolonged stimulation in virus-specific memory CD8+ T cells. In this study, the degree of cytotoxic capacity of virus-specific memory CD8+ T cells was directly proportional to the content of lytic molecules, which required antigenic stimulation over several days for maximal levels. This appeared to be modulated by increases in GrB transcription and microRNA-mediated posttranscriptional regulation of PRF expression. Clarifying the requirements for maximal cytotoxic capacity is critical to understanding how viral clearance might be mediated by memory cells and what functions should be induced by vaccines and immunotherapies.
Collapse
Affiliation(s)
- Stephen A Migueles
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Daniel C Rogan
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Noah V Gavil
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Elizabeth P Kelly
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sushila A Toulmin
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Lawrence T Wang
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Justin Lack
- NIAID Collaborative Bioinformatics Resource (NCBR), Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Maryland, USA
| | - Addison J Ward
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Patrick F Pryal
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Amanda K Ludwig
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Renata G Medina
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Benjamin J Apple
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Christina N Toumanios
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - April L Poole
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Catherine A Rehm
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sara E Jones
- Clinical Research Program Directorate, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Maryland, USA
| | - C Jason Liang
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Mark Connors
- HIV-Specific Immunity Section, Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
10
|
Abstract
The human liver is an organ with a diverse array of immunologic functions. Its unique anatomic position that leads to it receiving all the mesenteric venous blood, combined with its unique micro anatomy, allows it to serve as a sentinel for the body's immune system. Hepatocytes, biliary epithelial cells, Kupffer cells, stellate cells, and liver sinusoidal endothelial cells express key molecules that recruit and activate innate and adaptive immunity. Additionally, a diverse array of lymphoid and myeloid immune cells resides within and traffics to the liver in specific circumstances. Derangement of these trafficking mechanisms underlies the pathophysiology of autoimmune liver diseases, nonalcoholic steatohepatitis, and liver transplantation. Here, we review these pathways and interactions along with potential targets that have been identified to be exploited for therapeutic purposes.
Collapse
|
11
|
Suwandi JS, Laban S, Vass K, Joosten A, van Unen V, Lelieveldt BP, Höllt T, Zwaginga JJ, Nikolic T, Roep BO. Multidimensional analyses of proinsulin peptide-specific regulatory T cells induced by tolerogenic dendritic cells. J Autoimmun 2020; 107:102361. [DOI: 10.1016/j.jaut.2019.102361] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 11/06/2019] [Accepted: 11/06/2019] [Indexed: 12/18/2022]
|
12
|
Rv2626c and Rv2032 activate TH1 response and downregulate regulatory T cells in peripheral blood mononuclear cells of tuberculosis patients. Comp Immunol Microbiol Infect Dis 2019; 62:46-53. [DOI: 10.1016/j.cimid.2018.11.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 11/24/2018] [Accepted: 11/27/2018] [Indexed: 12/19/2022]
|
13
|
Seidel JA, Vukmanovic‐Stejic M, Muller‐Durovic B, Patel N, Fuentes‐Duculan J, Henson SM, Krueger JG, Rustin MHA, Nestle FO, Lacy KE, Akbar AN. Skin resident memory CD8 + T cells are phenotypically and functionally distinct from circulating populations and lack immediate cytotoxic function. Clin Exp Immunol 2018; 194:79-92. [PMID: 30030847 PMCID: PMC6156810 DOI: 10.1111/cei.13189] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2018] [Indexed: 02/06/2023] Open
Abstract
The in-depth understanding of skin resident memory CD8+ T lymphocytes (TRM ) may help to uncover strategies for their manipulation during disease. We investigated isolated TRM from healthy human skin, which expressed the residence marker CD69, and compared them to circulating CD8+ T cell populations from the same donors. There were significantly increased proportions of CD8+ CD45RA- CD27- T cells in the skin that expressed low levels of killer cell lectin-like receptor G1 (KLRG1), CD57, perforin and granzyme B. The CD8+ TRM in skin were therefore phenotypically distinct from circulating CD8+ CD45RA- CD27- T cells that expressed high levels of all these molecules. Nevertheless, the activation of CD8+ TRM with T cell receptor (TCR)/CD28 or interleukin (IL)-2 or IL-15 in vitro induced the expression of granzyme B. Blocking signalling through the inhibitory receptor programmed cell death 1 (PD)-1 further boosted granzyme B expression. A unique feature of some CD8+ TRM cells was their ability to secrete high levels of tumour necrosis factor (TNF)-α and IL-2, a cytokine combination that was not seen frequently in circulating CD8+ T cells. The cutaneous CD8+ TRM are therefore diverse, and appear to be phenotypically and functionally distinct from circulating cells. Indeed, the surface receptors used to distinguish differentiation stages of blood T cells cannot be applied to T cells in the skin. Furthermore, the function of cutaneous TRM appears to be stringently controlled by environmental signals in situ.
Collapse
Affiliation(s)
- J. A. Seidel
- Division of Infection and ImmunityUniversity College LondonUK
| | | | - B. Muller‐Durovic
- Division of Infection and ImmunityUniversity College LondonUK
- Department of BiomedicineUniversity of BaselBaselSwitzerland
| | - N. Patel
- Division of Infection and ImmunityUniversity College LondonUK
| | - J. Fuentes‐Duculan
- Laboratory for Investigative DermatologyThe Rockefeller UniversityNew YorkUSA
| | - S. M. Henson
- Division of Infection and ImmunityUniversity College LondonUK
- Present address:
William Harvey Research Institute Queen Mary University of LondonCharterhouse SquareLondon EC1M 6BQ
| | - J. G. Krueger
- Laboratory for Investigative DermatologyThe Rockefeller UniversityNew YorkUSA
| | | | - F. O. Nestle
- NIHR Biomedical Research Centre, Cutaneous Medicine and ImmunotherapySt John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, Guy’s Hospital, King’s College LondonLondonUK
| | - K. E. Lacy
- NIHR Biomedical Research Centre, Cutaneous Medicine and ImmunotherapySt John’s Institute of Dermatology, Division of Genetics and Molecular Medicine, Guy’s Hospital, King’s College LondonLondonUK
| | - A. N. Akbar
- Division of Infection and ImmunityUniversity College LondonUK
| |
Collapse
|
14
|
Ratajczak W, Niedźwiedzka-Rystwej P, Tokarz-Deptuła B, Deptuła W. Immunological memory cells. Cent Eur J Immunol 2018; 43:194-203. [PMID: 30135633 PMCID: PMC6102609 DOI: 10.5114/ceji.2018.77390] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 02/16/2018] [Indexed: 02/03/2023] Open
Abstract
This article reviews immunological memory cells, currently represented by T and B lymphocytes and natural killer (NK) cells, which determine a rapid and effective response against a second encounter with the same antigen. Among T lymphocytes, functions of memory cells are provided by their subsets: central memory, effector memory, tissue-resident memory, regulatory memory and stem memory T cells. Memory T and B lymphocytes have an essential role in the immunity against microbial pathogens but are also involved in autoimmunity and maternal-fetal tolerance. Furthermore, the evidence of immunological memory has been established for NK cells. NK cells can respond to haptens or viruses, which results in generation of antigen-specific memory cells. T, B and NK cells, which have a role in immunological memory, have been characterized phenotypically and functionally. During the secondary immune response, these cells are involved in the reaction against foreign antigens, including pathogens, and take part in autoimmune diseases, but also are crucial to immunological tolerance and vaccine therapy.
Collapse
Affiliation(s)
- Weronika Ratajczak
- Scientific Circle of Microbiologists, Faculty of Biology, University of Szczecin, Szczecin, Poland
| | | | - Beata Tokarz-Deptuła
- Department of Immunology, Faculty of Biology, University of Szczecin, Szczecin, Poland
| | - Wiesław Deptuła
- Department of Microbiology, Faculty of Biology, University of Szczecin, Szczecin, Poland
| |
Collapse
|
15
|
Brummelman J, Pilipow K, Lugli E. The Single-Cell Phenotypic Identity of Human CD8+ and CD4+ T Cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 341:63-124. [DOI: 10.1016/bs.ircmb.2018.05.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
16
|
Abstract
Epstein-Barr virus (EBV) is arguably one of the most successful pathogens of humans, persistently infecting over ninety percent of the world's population. Despite this high frequency of carriage, the virus causes apparently few adverse effects in the vast majority of infected individuals. Nevertheless, the potent growth transforming ability of EBV means the virus has the potential to cause malignancies in infected individuals. Indeed, EBV is thought to cause 1% of human malignancies, equating to 200,000 malignancies each year. A clear factor as to why virus-induced disease is relatively infrequent in healthy infected individuals is the presence of a potent immune response to EBV, in particular, that mediated by T cells. Thus, patient groups with immunodeficiencies or whose cellular immune response is suppressed have much higher frequencies of EBV-induced disease and, in at least some cases, these diseases can be controlled by restoration of the T-cell compartment. In this chapter, we will primarily review the role the αβ subset of T cells in the control of EBV in healthy and diseased individuals.
Collapse
Affiliation(s)
- Andrew D Hislop
- School of Cancer Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| | - Graham S Taylor
- School of Cancer Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
17
|
Wahid R, Fresnay S, Levine MM, Sztein MB. Immunization with Ty21a live oral typhoid vaccine elicits crossreactive multifunctional CD8+ T-cell responses against Salmonella enterica serovar Typhi, S. Paratyphi A, and S. Paratyphi B in humans. Mucosal Immunol 2015; 8:1349-59. [PMID: 25872480 PMCID: PMC4607552 DOI: 10.1038/mi.2015.24] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 02/06/2015] [Indexed: 02/04/2023]
Abstract
Previously we have extensively characterized Salmonella enterica serovar Typhi (S. Typhi)-specific cell-mediated immune (CMI) responses in volunteers orally immunized with the licensed Ty21a typhoid vaccine. In this study we measured Salmonella-specific multifunctional (MF) CD8+ T-cell responses to further investigate whether Ty21a elicits crossreactive CMI against S. Paratyphi A and S. Paratyphi B that also cause enteric fever. Ty21a-elicited crossreactive CMI responses against all three Salmonella serotypes were predominantly observed in CD8+ T effector/memory (T(EM)) and, to a lesser extent, in CD8+CD45RA+ T(EM) (T(EMRA)) subsets. These CD8+ T-cell responses were largely mediated by MF cells coproducing interferon-γ and macrophage inflammatory protein-1β and expressing CD107a with or without tumor necrosis factor-α. Significant proportions of Salmonella-specific MF cells expressed the gut-homing molecule integrin α4β7. In most subjects, similar MF responses were observed to S. Typhi and S. Paratyphi B, but not to S. Paratyphi A. These results suggest that Ty21a elicits MF CMI responses against Salmonella that could be critical in clearing the infection. Moreover, because S. Paratyphi A is a major public concern and Ty21a was shown in field studies not to afford cross-protection to S. Paratyphi A, these results will be important in developing a S. Typhi/S. Paratyphi A bivalent vaccine against enteric fevers.
Collapse
Affiliation(s)
- Rezwanul Wahid
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Stephanie Fresnay
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Myron M. Levine
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Marcelo B. Sztein
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
18
|
Vieira Braga FA, Hertoghs KML, van Lier RAW, van Gisbergen KPJM. Molecular characterization of HCMV-specific immune responses: Parallels between CD8(+) T cells, CD4(+) T cells, and NK cells. Eur J Immunol 2015; 45:2433-45. [PMID: 26228786 DOI: 10.1002/eji.201545495] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 07/15/2015] [Accepted: 07/28/2015] [Indexed: 11/07/2022]
Abstract
CD8(+) T cells are important for immunity against human cytomegalovirus (HCMV). The HCMV-specific CD8(+) T-cell response is characterized by the accumulation of terminally differentiated effector cells that have downregulated the costimulatory molecules CD27 and CD28. These HCMV-specific CD8(+) T cells maintain high levels of cytotoxic molecules such as granzyme B and rapidly produce the inflammatory cytokine IFN-γ upon activation. Remarkably, HCMV-specific CD8(+) T cells are able to persist long term as fully functional effector cells, suggesting a unique differentiation pathway that is distinct from the formation of memory CD8(+) T cells after infection with acute viruses. In this review, we aim to highlight the most recent developments in HCMV-specific CD8(+) T-cell differentiation, maintenance, tissue distribution, metabolism and function. HCMV also induces the differentiation of effector CD4(+) T cells and NK cells, which share characteristics with HCMV-specific CD8(+) T cells. We propose that the overlap in differentiation of NK cells, CD4(+) and CD8(+) T cells after HCMV infection may be regulated by a shared transcriptional machinery. A better understanding of the molecular framework of HCMV-specific CD8(+) T-cell responses may benefit vaccine design, as these cells uniquely combine the capacity to rapidly respond to infection with long-term survival.
Collapse
Affiliation(s)
- Felipe A Vieira Braga
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory AMC/UvA, Amsterdam, The Netherlands
| | - Kirsten M L Hertoghs
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory AMC/UvA, Amsterdam, The Netherlands
| | - René A W van Lier
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory AMC/UvA, Amsterdam, The Netherlands
| | - Klaas P J M van Gisbergen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory AMC/UvA, Amsterdam, The Netherlands
| |
Collapse
|
19
|
Success and failure of the cellular immune response against HIV-1. Nat Immunol 2015; 16:563-70. [PMID: 25988888 DOI: 10.1038/ni.3161] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 03/27/2015] [Indexed: 02/07/2023]
Abstract
The cellular immune response to HIV-1 has now been studied in extraordinary detail. A very large body of data provides the most likely reasons that the HIV-specific cellular immune response succeeds in a small number of people but fails in most. Understanding the success and failure of the HIV-specific cellular immune response has implications that extend not only to immunotherapies and vaccines for HIV-1 but also to the cellular immune response in other disease states. This Review focuses on the mechanisms that are most likely responsible for durable and potent immunologic control of HIV-1. Although we now have a detailed picture of the cellular immune responses to HIV-1, important questions remain regarding the nature of these responses and how they arise.
Collapse
|
20
|
Nakae Y, Oka Y, Fujiki F, Morimoto S, Kamiya T, Takashima S, Nakata J, Nishida S, Nakajima H, Hosen N, Tsuboi A, Kyo T, Oji Y, Mizuguchi K, Kumanogoh A, Sugiyama H. Two distinct effector memory cell populations of WT1 (Wilms' tumor gene 1)-specific cytotoxic T lymphocytes in acute myeloid leukemia patients. Cancer Immunol Immunother 2015; 64:791-804. [PMID: 25835542 PMCID: PMC11028643 DOI: 10.1007/s00262-015-1683-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 03/14/2015] [Indexed: 10/23/2022]
Abstract
Wilms' tumor gene 1 (WT1) protein is a promising tumor-associated antigen for cancer immunotherapy. We have been performing WT1 peptide vaccination with good clinical responses in over 750 patients with leukemia or solid cancers. In this study, we generated single-cell gene-expression profiles of the effector memory (EM) subset of WT1-specific cytotoxic T lymphocytes (CTLs) in peripheral blood of nine acute myeloid leukemia patients treated with WT1 peptide vaccine, in order to discriminate responders (WT1 mRNA levels in peripheral blood decreased to undetectable levels, decreased but stayed at abnormal levels, were stable at undetectable levels, or remained unchanged from the initial abnormal levels more than 6 months after WT1 vaccination) from non-responders (leukemic blast cells and/or WT1 mRNA levels increased relative to the initial state within 6 months of WT1 vaccination) prior to WT1 vaccination. Cluster and principal component analyses performed using 83 genes did not discriminate between responders and non-responders prior to WT1 vaccination. However, these analyses revealed that EM subset of WT1-specific CTLs could be divided into two groups: the "activated" and "quiescent" states; in responders, EM subset of the CTLs shifted to the "quiescent" state, whereas in non-responders, those shifted to the "activated" state following WT1 vaccination. These results demonstrate for the first time the existence of two distinct EM states, each of which was characteristic of responders or non-responders, of WT1-specific CTLs in AML patients, and raises the possibility of using advanced gene-expression profile analysis to clearly discriminate between responders and non-responders prior to WT1 vaccination.
Collapse
MESH Headings
- Adult
- Aged
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Female
- Gene Expression Profiling
- Humans
- Immunologic Memory/immunology
- Immunotherapy/methods
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/therapy
- Male
- Middle Aged
- Principal Component Analysis
- RNA, Messenger/blood
- RNA, Messenger/genetics
- T-Lymphocytes, Cytotoxic/cytology
- T-Lymphocytes, Cytotoxic/immunology
- Vaccines, Subunit/immunology
- Vaccines, Subunit/therapeutic use
- WT1 Proteins/genetics
- WT1 Proteins/immunology
Collapse
Affiliation(s)
- Yoshiki Nakae
- Departments of Respiratory Medicine, Allergy and Rheumatic Diseases, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Yoshihiro Oka
- Departments of Respiratory Medicine, Allergy and Rheumatic Diseases, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871 Japan
- Department of Cancer Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871 Japan
- Department of Immunopathology, Immunology Frontier Research Center (World Premier International Research Center), Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Fumihiro Fujiki
- Department of Cancer Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Soyoko Morimoto
- Department of Cancer Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Toshio Kamiya
- Department of Functional Diagnostic Science, Graduate School of Medicine, Osaka University, 1-7 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Satoshi Takashima
- Departments of Respiratory Medicine, Allergy and Rheumatic Diseases, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Jun Nakata
- Department of Cancer Immunotherapy, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Sumiyuki Nishida
- Departments of Respiratory Medicine, Allergy and Rheumatic Diseases, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Hiroko Nakajima
- Department of Cancer Immunology, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Naoki Hosen
- Department of Cancer Stem Cell Biology, Graduate School of Medicine, Osaka University, 1-7 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Akihiro Tsuboi
- Department of Cancer Immunotherapy, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Taiichi Kyo
- Department of Hematology, Hiroshima Red Cross and Atomic Bomb Survivor Hospital, 1-9-6 Sendamachi, Nakaku, Hiroshima-City, Hiroshima 730-0052 Japan
| | - Yusuke Oji
- Department of Cancer Stem Cell Biology, Graduate School of Medicine, Osaka University, 1-7 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Kenji Mizuguchi
- National Institute of Biomedical Innovation, 7-6-8 Saitoasagi, Ibaraki, Osaka 567-0085 Japan
| | - Atsushi Kumanogoh
- Departments of Respiratory Medicine, Allergy and Rheumatic Diseases, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871 Japan
- Department of Immunopathology, Immunology Frontier Research Center (World Premier International Research Center), Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871 Japan
| | - Haruo Sugiyama
- Department of Functional Diagnostic Science, Graduate School of Medicine, Osaka University, 1-7 Yamadaoka, Suita, Osaka 565-0871 Japan
| |
Collapse
|
21
|
Shook DR, Triplett BM, Eldridge PW, Kang G, Srinivasan A, Leung W. Haploidentical stem cell transplantation augmented by CD45RA negative lymphocytes provides rapid engraftment and excellent tolerability. Pediatr Blood Cancer 2015; 62:666-73. [PMID: 25559618 DOI: 10.1002/pbc.25352] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 10/20/2014] [Indexed: 12/31/2022]
Abstract
BACKGROUND Haploidentical donors are being increasingly used for allogeneic hematopoietic cell transplantation (HCT). However, the requisite T-cell depletion results in a profound and often long-lasting immunocompromised state, and donor lymphocyte infusions bring a risk of graft-versus-host disease (GVHD). Naïve T-cells are believed to be among the most alloreactive T-cell subset and can be identified by CD45RA expression. Allogeneic HCT using CD45RA depletion has not been previously described for haploidentical donors. PROCEDURE Eight children with relapsed or refractory solid tumors were transplanted following myeloablative conditioning. Each patient received two cell products, one created by CD3 depletion and the other through CD45RA depletion. RESULTS Median CD34 recovery was 59.2% with CD45RA depletion, compared to 82.4% using CD3 depletion. Median CD3+ T-cell dose after CD45RA reduction was 99.2 × 10(6) cells/kg, yet depletion of CD3+ CD45RA+ cells exceeded 4.5 log. CD45RA depletion also resulted in substantial depletion of B-cells (median 2.45 log). All eight patients engrafted within 14 days and rapidly achieved 100% donor chimerism. No acute GVHD or secondary graft failure was observed. CONCLUSIONS CD45RA depletion is a novel approach to haploidentical HCT that offers rapid engraftment with minimal risk of GVHD.
Collapse
Affiliation(s)
- David R Shook
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee 38105; Department of Pediatrics, University of Tennessee Health Science Center, College of Medicine, Memphis, Tennessee 38163
| | | | | | | | | | | |
Collapse
|
22
|
Mbow M, de Jong SE, Meurs L, Mboup S, Dieye TN, Polman K, Yazdanbakhsh M. Changes in immunological profile as a function of urbanization and lifestyle. Immunology 2015; 143:569-77. [PMID: 24924958 DOI: 10.1111/imm.12335] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 05/14/2014] [Accepted: 06/09/2014] [Indexed: 12/12/2022] Open
Abstract
Differences in lifestyle and break with natural environment appear to be associated with changes in the immune system resulting in various adverse health effects. Although genetics can have a major impact on the immune system and disease susceptibility, the contribution of environmental factors is thought to be substantial. Here, we investigated the immunological profile of healthy volunteers living in a rural and an urban area of a developing African country (Senegal), and in a European country (the Netherlands). Using flow cytometry, we investigated T helper type 1 (Th1), Th2, Th17, Th22 and regulatory T cells, as well as CD4(+) T-cell and B-cell activation markers, and subsets of memory T and B cells in the peripheral blood. Rural Senegalese had significantly higher frequencies of Th1, Th2 and Th22 cells, memory CD4(+) T and B cells, as well as activated CD4(+) T and B cells compared with urban Senegalese and urban Dutch people. Within the Senegalese population, rural paritcipants displayed significantly higher frequencies of Th2 and Th22 cells, as well as higher pro-inflammatory and T-cell activation and memory profiles compared with the urban population. The greater magnitude of immune activation and the enlarged memory pool, together with Th2 polarization, seen in rural participants from Africa, followed by urban Africans and Europeans suggest that environmental changes may define immunological footprints, which could have consequences for disease patterns in general and vaccine responses in particular.
Collapse
Affiliation(s)
- Moustapha Mbow
- Immunology Department of the Laboratory of Bacteriology and Virology of Aristide Le Dantec University Hospital, Dakar, Senegal; Leiden Immunoparasitology Group, Department of Parasitology, Leiden University Medical Centre, Leiden, the Netherlands; Department of Biomedical Sciences of the Institute of Tropical Medicine of Antwerp, Antwerp, Belgium
| | | | | | | | | | | | | |
Collapse
|
23
|
Williams GP, Pachnio A, Long HM, Rauz S, Curnow SJ. Cytokine production and antigen recognition by human mucosal homing conjunctival effector memory CD8+ T cells. Invest Ophthalmol Vis Sci 2014; 55:8523-30. [PMID: 25395484 DOI: 10.1167/iovs.14-15133] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
PURPOSE Conjunctival epithelial T cells are dominated by CD3(+)CD56-TCRαβ(+)CD8αβ(+) lymphocytes. In this study we explored the antigen experience status, mucosal homing phenotype, cytokine expression, and viral antigen recognition of conjunctival epithelial CD8(+) T cells from healthy individuals. METHODS Following ocular surface impression cytology, conjunctival cells were recovered by gentle agitation and analyzed by flow cytometry for cell surface markers, cytokine production (stimulated by phorbol 12-myristate 13-acetate [PMA]/ionomycin), and Epstein-Barr virus (EBV)/cytomegalovirus (CMV) immunodominant epitope recognition using major histocompatibility complex (MHC) class I peptide tetramers. RESULTS In contrast to peripheral blood, conjunctival epithelial CD8(+) T cells were dominantly CD45RA(-)CCR7(-) effector memory cells, and the vast majority expressed the mucosal homing integrin αEβ7. Conjunctival memory CD8(+) T cells maintained effector functions with the ability to secrete IFN-γ and expression of Granzyme B, although they expressed significantly reduced amounts per cell compared to peripheral blood T cells. Interestingly, herpetic virus-specific CD8(+) T cells recognizing epitopes derived from EBV and CMV could be detected in the conjunctival cells of healthy virus carriers, although they were generally at lower frequencies than in the peripheral blood of the same donor. Virus-specific conjunctival CD8(+) T cells were dominated by CD45RA(-)CCR7(-) effector memory cells that expressed αEβ7. CONCLUSIONS These data demonstrate that the majority of conjunctival epithelial CD8(+) T cells are mucosal homing αEβ7(+) effector memory T cells, which can recognize viral epitopes and are capable of secreting Granzyme B and IFN-γ.
Collapse
Affiliation(s)
- Geraint P Williams
- Academic Unit of Ophthalmology, Birmingham and Midland Eye Centre, City Hospital, Birmingham, School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom Centre for Translational Inflammation Research, Queen Elizabeth Hospital Birmingham, School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Annette Pachnio
- School of Cancer Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Heather M Long
- School of Cancer Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Saaeha Rauz
- Academic Unit of Ophthalmology, Birmingham and Midland Eye Centre, City Hospital, Birmingham, School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom Centre for Translational Inflammation Research, Queen Elizabeth Hospital Birmingham, School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - S John Curnow
- Academic Unit of Ophthalmology, Birmingham and Midland Eye Centre, City Hospital, Birmingham, School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom Centre for Translational Inflammation Research, Queen Elizabeth Hospital Birmingham, School of Immunity and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
24
|
Henson SM, Lanna A, Riddell NE, Franzese O, Macaulay R, Griffiths SJ, Puleston DJ, Watson AS, Simon AK, Tooze SA, Akbar AN. p38 signaling inhibits mTORC1-independent autophagy in senescent human CD8⁺ T cells. J Clin Invest 2014; 124:4004-16. [PMID: 25083993 PMCID: PMC4151208 DOI: 10.1172/jci75051] [Citation(s) in RCA: 289] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Accepted: 06/13/2014] [Indexed: 01/09/2023] Open
Abstract
T cell senescence is thought to contribute to immune function decline, but the pathways that mediate senescence in these cells are not clear. Here, we evaluated T cell populations from healthy volunteers and determined that human CD8+ effector memory T cells that reexpress the naive T cell marker CD45RA have many characteristics of cellular senescence, including decreased proliferation, defective mitochondrial function, and elevated levels of both ROS and p38 MAPK. Despite their apparent senescent state, we determined that these cells secreted high levels of both TNF-α and IFN-γ and showed potent cytotoxic activity. We found that the senescent CD45RA-expressing population engaged anaerobic glycolysis to generate energy for effector functions. Furthermore, inhibition of p38 MAPK signaling in senescent CD8+ T cells increased their proliferation, telomerase activity, mitochondrial biogenesis, and fitness; however, the extra energy required for these processes did not arise from increased glucose uptake or oxidative phosphorylation. Instead, p38 MAPK blockade in these senescent cells induced an increase in autophagy through enhanced interactions between p38 interacting protein (p38IP) and autophagy protein 9 (ATG9) in an mTOR-independent manner. Together, our findings describe fundamental metabolic requirements of senescent primary human CD8+ T cells and demonstrate that p38 MAPK blockade reverses senescence via an mTOR-independent pathway.
Collapse
Affiliation(s)
- Sian M. Henson
- Division of Infection and Immunity, University College London, London, United Kingdom. Department of Systems Medicine, University of Tor Vergata, Rome, Italy. MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom. Secretory Pathways Laboratory, London Research Institute, Cancer Research UK, London, United Kingdom
| | - Alessio Lanna
- Division of Infection and Immunity, University College London, London, United Kingdom. Department of Systems Medicine, University of Tor Vergata, Rome, Italy. MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom. Secretory Pathways Laboratory, London Research Institute, Cancer Research UK, London, United Kingdom
| | - Natalie E. Riddell
- Division of Infection and Immunity, University College London, London, United Kingdom. Department of Systems Medicine, University of Tor Vergata, Rome, Italy. MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom. Secretory Pathways Laboratory, London Research Institute, Cancer Research UK, London, United Kingdom
| | - Ornella Franzese
- Division of Infection and Immunity, University College London, London, United Kingdom. Department of Systems Medicine, University of Tor Vergata, Rome, Italy. MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom. Secretory Pathways Laboratory, London Research Institute, Cancer Research UK, London, United Kingdom
| | - Richard Macaulay
- Division of Infection and Immunity, University College London, London, United Kingdom. Department of Systems Medicine, University of Tor Vergata, Rome, Italy. MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom. Secretory Pathways Laboratory, London Research Institute, Cancer Research UK, London, United Kingdom
| | - Stephen J. Griffiths
- Division of Infection and Immunity, University College London, London, United Kingdom. Department of Systems Medicine, University of Tor Vergata, Rome, Italy. MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom. Secretory Pathways Laboratory, London Research Institute, Cancer Research UK, London, United Kingdom
| | - Daniel J. Puleston
- Division of Infection and Immunity, University College London, London, United Kingdom. Department of Systems Medicine, University of Tor Vergata, Rome, Italy. MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom. Secretory Pathways Laboratory, London Research Institute, Cancer Research UK, London, United Kingdom
| | - Alexander Scarth Watson
- Division of Infection and Immunity, University College London, London, United Kingdom. Department of Systems Medicine, University of Tor Vergata, Rome, Italy. MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom. Secretory Pathways Laboratory, London Research Institute, Cancer Research UK, London, United Kingdom
| | - Anna Katharina Simon
- Division of Infection and Immunity, University College London, London, United Kingdom. Department of Systems Medicine, University of Tor Vergata, Rome, Italy. MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom. Secretory Pathways Laboratory, London Research Institute, Cancer Research UK, London, United Kingdom
| | - Sharon A. Tooze
- Division of Infection and Immunity, University College London, London, United Kingdom. Department of Systems Medicine, University of Tor Vergata, Rome, Italy. MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom. Secretory Pathways Laboratory, London Research Institute, Cancer Research UK, London, United Kingdom
| | - Arne N. Akbar
- Division of Infection and Immunity, University College London, London, United Kingdom. Department of Systems Medicine, University of Tor Vergata, Rome, Italy. MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom. Secretory Pathways Laboratory, London Research Institute, Cancer Research UK, London, United Kingdom
| |
Collapse
|
25
|
Wheat R, Roberts C, Waterboer T, Steele J, Marsden J, Steven NM, Blackbourn DJ. Inflammatory cell distribution in primary merkel cell carcinoma. Cancers (Basel) 2014; 6:1047-64. [PMID: 24961933 PMCID: PMC4074816 DOI: 10.3390/cancers6021047] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 03/28/2014] [Accepted: 03/31/2014] [Indexed: 12/11/2022] Open
Abstract
Merkel cell carcinoma (MCC) is an aggressive poorly differentiated neuroendocrine cutaneous carcinoma associated with older age, immunodeficiency and Merkel cell polyomavirus (MCPyV) integrated within malignant cells. The presence of intra-tumoural CD8+ lymphocytes reportedly predicts better MCC-specific survival. In this study, the distribution of inflammatory cells and properties of CD8+ T lymphocytes within 20 primary MCC specimens were characterised using immunohistochemistry and multicolour immunofluorescent staining coupled to confocal microscopy. CD8+ cells and CD68+ macrophages were identified in 19/20 primary MCC. CD20+ B cells were present in 5/10, CD4+ cells in 10/10 and FoxP3+ cells in 7/10 specimens. Only two specimens had almost no inflammatory cells. Within specimens, inflammatory cells followed the same patchy distribution, focused at the edge of sheets and nodules and, in some cases, more intense in trabecular areas. CD8+ cells were outside vessels on the edge of tumour. Those few within malignant sheets typically lined up in fine septa not contacting MCC cells expressing MCPyV large T antigen. The homeostatic chemokine CXCL12 was expressed outside malignant nodules whereas its receptor CXCR4 was identified within tumour but not on CD8+ cells. CD8+ cells lacked CXCR3 and granzyme B expression irrespective of location within stroma versus malignant nodules or of the intensity of the intra-tumoural infiltrate. In summary, diverse inflammatory cells were organised around the margin of malignant deposits suggesting response to aberrant signaling, but were unable to penetrate the tumour microenvironment itself to enable an immune response against malignant cells or their polyomavirus.
Collapse
Affiliation(s)
- Rachel Wheat
- School of Cancer Sciences and CR UK Centre for Cancer Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK.
| | - Claudia Roberts
- School of Cancer Sciences and CR UK Centre for Cancer Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK.
| | - Tim Waterboer
- Infection and Cancer Program, DKFZ (German Cancer Research Centre), 69120 Heidelberg, Germany.
| | - Jane Steele
- Human Biomaterials Resource Centre, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK.
| | - Jerry Marsden
- University Hospitals Birmingham NHS Foundation Trust, New Queen Elizabeth Hospital Birmingham, Mindelsohn Way, Edgbaston, Birmingham, B15 2WB, UK.
| | - Neil M Steven
- School of Cancer Sciences and CR UK Centre for Cancer Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK.
| | - David J Blackbourn
- Department of Microbial and Cellular Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford, Surrey, GU2 7XH, UK.
| |
Collapse
|
26
|
Engineering human peripheral blood stem cell grafts that are depleted of naïve T cells and retain functional pathogen-specific memory T cells. Biol Blood Marrow Transplant 2014; 20:705-16. [PMID: 24525279 DOI: 10.1016/j.bbmt.2014.01.032] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Accepted: 01/29/2014] [Indexed: 12/14/2022]
Abstract
Graft-versus-host disease (GVHD) is a frequent major complication of allogeneic hematopoietic cell transplantation (HCT). Approaches that selectively deplete T cells that cause GVHD from allogeneic stem cell grafts and preserve T cells specific for pathogens may improve HCT outcomes. It has been hypothesized that the majority of T cells that can cause GVHD reside within the naïve T cell (TN) subset, and previous studies performed in mouse models and with human cells in vitro support this hypothesis. As a prelude to translating these findings to the clinic, we developed and evaluated a novel 2-step clinically compliant procedure for manipulating peripheral blood stem cells (PBSC) to remove TN, preserve CD34(+) hematopoietic stem cells, and provide for a fixed dose of memory T cells (TM) that includes T cells with specificity for common opportunistic pathogens encountered after HCT. Our studies demonstrate effective and reproducible performance of the immunomagnetic cell selection procedure for depleting TN. Moreover, after cell processing, the CD45RA-depleted PBSC products are enriched for CD4(+) and CD8(+) TM with a central memory phenotype and contain TM cells that are capable of proliferating and producing effector cytokines in response to opportunistic pathogens.
Collapse
|
27
|
Derhovanessian E, Maier AB, Hähnel K, Zelba H, de Craen AJM, Roelofs H, Slagboom EP, Westendorp RGJ, Pawelec G. Lower proportion of naïve peripheral CD8+ T cells and an unopposed pro-inflammatory response to human Cytomegalovirus proteins in vitro are associated with longer survival in very elderly people. AGE (DORDRECHT, NETHERLANDS) 2013; 35:1387-1399. [PMID: 22661297 PMCID: PMC3705124 DOI: 10.1007/s11357-012-9425-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 04/30/2012] [Indexed: 06/01/2023]
Abstract
The low percentages of naïve T cells commonly observed in elderly people are thought to be causally associated with mortality, primarily from infectious disease, and are taken as a hallmark of "immunosenescence". Whether low levels of naive cells actually do associate with mortality has, however, not been tested in longitudinal studies. Here, we present correlations between peripheral T-cell phenotypes and 8-year survival in individuals from the population-based prospective Leiden 85-plus Study. Counter-intuitively, we found that a lower frequency of naïve CD8+ T cells (characterized as CD45RA+CCR7+CD27+CD28+) at baseline (>88 years) correlated with significantly better survival, while there was a tendency for the reciprocal accumulation of late-differentiated effector memory cells (CD45RA-CCR7-CD27-CD28-) also to associate with better survival. These findings suggest that better retention of memory cells specific for previously encountered antigens may provide a survival advantage in this particular population. Given the prevalence of Cytomegalovirus (CMV) and its reported association with immunosenescence, we tested whether memory for this potential pathogen was relevant to survival. We found that individuals mounting an exclusively pro-inflammatory ex vivo response (TNF, IFN-γ, IL-17) to the major CMV target molecules pp65 and IE1 had a significant survival advantage over those also having anti-inflammatory responses (IL-10). These findings suggest that higher levels of naïve T cells may not necessarily be associated with a survival advantage and imply that the nature of immunosurveillance against CMV may be crucial for remaining longevity, at least in the very elderly.
Collapse
Affiliation(s)
- Evelyna Derhovanessian
- Department of Internal Medicine II, Centre for Medical Research, University of Tübingen, Waldhörnlestrasse 22, 72072, Tübingen, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Moreno S, Alvarez B, Martínez P, Uenishi H, Revilla C, Ezquerra A, Alonso F, Domínguez J. Analysis of chemokine receptor CCR7 expression on porcine blood T lymphocytes using a CCL19-Fc fusion protein. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2013; 39:207-213. [PMID: 23219903 DOI: 10.1016/j.dci.2012.11.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Revised: 11/21/2012] [Accepted: 11/21/2012] [Indexed: 06/01/2023]
Abstract
The chemokine receptor CCR7 has been a useful marker for the characterization of human and mouse T cell subsets. We have produced the porcine CCR7 ligand CCL19 fused to the human IgG1 Fc fragment, and used it to analyse CCR7 expression in swine. CCL19-Fc bound to and induced the migration of cells expressing porcine CCR7 but not of untransfected cells, corroborating its specificity. On blood lymphocytes, CCL19-Fc labelled the majority of CD4(+) T cells expressing the 2E3 marker, associated with a naïve phenotype, whereas the 2E3(-) cells were mostly negative. Among CD8(+) T cells CCL19-Fc labelled two subsets: one, CD8β(hi) CD11a(lo) CD45RA(+), perforin(-/lo) , which produced low amounts of IFN-γ after stimulation, which might correspond to naïve cells; and a second small population of CD8β(lo) cells which expressed high levels of CD11a, and were mostly CD45RA(-), a phenotype which resembles that of human central memory T cells.
Collapse
Affiliation(s)
- S Moreno
- Dpto. de Biotecnología, INIA, Ctra de la Coruña Km 7.5, 28040 Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Inhibitory potential of subpopulations of CD8+ T cells in HIV-1-infected elite suppressors. J Virol 2012; 86:13679-88. [PMID: 23055552 DOI: 10.1128/jvi.02439-12] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Elite controllers or suppressors (ES) are HIV-1-infected individuals who suppress viral replication to clinically undetectable levels without antiretroviral therapy. Understanding the mechanisms by which ES control viral replication may prove informative for the design of a therapeutic vaccine. Qualitative differences in the CD8(+) T cell response have been implicated in control. Therefore, we isolated CD8(+) T cells from ES and characterized the ability of sorted memory and activation subpopulations to control viral replication at various effector-to-target cell ratios using a novel modification of a CD8(+) T cell suppression assay. The effector memory and terminal effector subpopulations of memory CD8(+) T cells had the highest inhibitory potential over the course of a 3-day in vitro infection. Interestingly, after 5 days of infection, central memory CD8(+) T cells were also very effective at suppressing viral replication. No significant correlation between the suppression of viral replication and the number of HIV-1-specific CD8(+) T cells was observed. HLA-DR(-) CD38(+) CD8(+) T cells possessed the lowest inhibitory potential of the activation subpopulations. Taken together, our data suggest that there are key differences in the magnitude and kinetics of the suppression of HIV-1 replication by different CD8(+) T cell subsets. These data should guide the development of an effective, cellular therapeutic vaccine that has the potential to elicit similar CD8(+) T cell responses.
Collapse
|
30
|
Qualitative host factors associated with immunological control of HIV infection by CD8 T cells. Curr Opin HIV AIDS 2012; 1:28-33. [PMID: 19372780 DOI: 10.1097/01.coh.0000194108.14601.69] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Despite significant technical advances that have permitted an increasingly more quantitative and detailed study of virus-specific cellular immunity over the past few years, our understanding of the nature of immunological control in rare cases of non-progressive HIV infection and diminished control in the majority of untreated chronically infected patients remains incomplete. This review will summarize recent findings and points of controversy within areas of active investigation of the cellular immune response to HIV. RECENT FINDINGS It is now appreciated that high frequencies of virus-specific CD8 T cells are readily detectable in chronic HIV infection, but do not restrict viral replication. For this reason, attention has shifted to qualitative features of the host immune response that might accurately determine the restriction of viral replication. A number of qualitative changes in the phenotype, cytokine secretion, and proliferative capacity of HIV-specific CD8 T cells of progressors have recently been described. SUMMARY Given that the desired response to the majority of vaccines in pre-clinical or clinical testing is to stimulate cellular immunity in an attempt to alter disease progression, understanding these qualitative features is of particular relevance. Further study will probably yield critical information for the means to stimulate effective immunity in vaccinees, prevent the loss of control of viral replication upon infection of vaccinees, or induce durable immunological control in humans already infected with HIV.
Collapse
|
31
|
Properties of end-stage human T cells defined by CD45RA re-expression. Curr Opin Immunol 2012; 24:476-81. [PMID: 22554789 DOI: 10.1016/j.coi.2012.04.001] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 04/08/2012] [Indexed: 01/10/2023]
Abstract
Persistent viral infections, inflammatory syndromes and ageing all induce the accumulation of highly differentiated CD45RA re-expressing memory T cells. These cells increase during ageing, especially in individuals who are infected with cytomegalovirus (CMV). These cells have decreased proliferative capacity, increased activation of senescence signalling pathways and greater susceptibility to apoptosis in vitro. However these cells are capable of multiple effector functions and thus bear all the hallmarks of short-lived effector T cells. This indicates that senescence signalling may govern the unique characteristics of effector T cells. In this article, we address the functional and migratory properties of these T cells and mechanisms that are involved in their generation. Finally we assess the potential for manipulation of their activity and whether this may improve immune function during ageing.
Collapse
|
32
|
McAlpine SM, Issekutz TB, Marshall JS. Virus stimulation of human mast cells results in the recruitment of CD56⁺ T cells by a mechanism dependent on CCR5 ligands. FASEB J 2011; 26:1280-9. [PMID: 22125314 DOI: 10.1096/fj.11-188979] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The trafficking of effector cells to sites of infection is crucial for antiviral responses. However, the mechanisms of recruitment of the interferon-γ-producing and cytotoxic CD56(+) T cells are poorly understood. Human mast cells are sentinel cells found in the skin and airway and produce selected proinflammatory mediators in response to multiple pathogen-associated signals. The role of human mast cell-derived chemokines in T-cell recruitment to virus infection was examined. Supernatants from primary human cord blood-derived mast cells (CBMCs) infected with mammalian reovirus were examined for chemokine production and utilized in chemotaxis assays. Virus-infected CBMCs produced several chemokines, including CCL3, CCL4, and CCL5. Supernatants from reovirus-infected CBMCs selectively induced the chemotaxis of CD8(+) T cells (10±1%) and CD3(+)CD56(+) T cells (19±5%). CD56(+) T-cell migration was inhibited by pertussis toxin (65±9%) and met-RANTES (56±7%), a CCR1/CCR5 antagonist. CD56(+) T cells expressed CCR5, but little CCR1. The depletion of CCL3, CCL4, and CCL5 from reovirus-infected CBMC supernatants significantly (41±10%) inhibited CD56(+) T-cell chemotaxis. This study demonstrates a novel role for mast cells and CCR5 in CD56(+) T-cell trafficking and suggests that human mast cells enhance immunity to viruses through the selective recruitment of cytotoxic effector cells to virus infection sites. These findings could be exploited to enhance local T-cell responses in chronic viral infection and malignancies at mast cell-rich sites.
Collapse
Affiliation(s)
- Sarah M McAlpine
- Dalhousie Inflammation Group, Dalhousie University, 5850 College St., Halifax, NS B3H 1X5, Canada
| | | | | |
Collapse
|
33
|
Di Mitri D, Azevedo RI, Henson SM, Libri V, Riddell NE, Macaulay R, Kipling D, Soares MVD, Battistini L, Akbar AN. Reversible Senescence in Human CD4+CD45RA+CD27− Memory T Cells. THE JOURNAL OF IMMUNOLOGY 2011; 187:2093-100. [DOI: 10.4049/jimmunol.1100978] [Citation(s) in RCA: 165] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
34
|
Makedonas G, Betts MR. Living in a house of cards: re-evaluating CD8+ T-cell immune correlates against HIV. Immunol Rev 2011; 239:109-24. [PMID: 21198668 DOI: 10.1111/j.1600-065x.2010.00968.x] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The Merck STEP and the Thai RV144 human immunodeficiency virus (HIV) vaccine trials confirmed that we still have a long way to go before developing a prophylactic HIV vaccine. The main issue at hand is that we have yet to identify an immunological correlate of protection against HIV. While many question the T-cell-based approach towards vaccine development, it is likely that T cells will be a necessary part of any vaccine strategy. CD8(+) T cells remain an attractive option because of their ability to specifically recognize and eliminate virally infected host cells. In this review, we recapitulate the evidence for CD8(+) T cells as an immunological correlate against HIV, but more importantly, we assess the means by which we evaluate their antiviral capacity. To achieve a breakthrough in the domain of T-cell-based HIV vaccine development, it has become abundantly clear that we must overhaul our system of immune monitoring and come up with a 'rational' tactic to evaluate the efficacy of HIV-specific CD8(+) T cells.
Collapse
|
35
|
Siena L, Gjomarkaj M, Elliot J, Pace E, Bruno A, Baraldo S, Saetta M, Bonsignore MR, James A. Reduced apoptosis of CD8+ T-lymphocytes in the airways of smokers with mild/moderate COPD. Respir Med 2011; 105:1491-500. [PMID: 21612902 DOI: 10.1016/j.rmed.2011.04.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Revised: 03/16/2011] [Accepted: 04/25/2011] [Indexed: 10/18/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is characterised by chronic inflammation in airways and lung parenchyma. CD8+ T-lymphocytes, crucial effector and regulatory cells in inflammation, are increased in the central and peripheral airways in COPD. The aim of this study was to assess the role of apoptosis in the accumulation of CD8+ T-lymphocytes within the airway wall in COPD. We examined the submucosa of transverse sections of central and peripheral airways from post-operative tissues from non-smokers (n = 16), smokers with normal lung function (n = 16), smokers with mild/moderate COPD (n = 16), and smokers with severe/very severe COPD (n = 9). TUNEL and immunohistochemistry techniques were used to identify apoptosis and cell phenotype, respectively. The percentage of apoptotic CD8+ T-lymphocytes was significantly lower (p < 0.0001) in smokers with mild/moderate COPD than in non-smokers, smokers with normal lung function, and smokers with severe/very severe COPD, and was positively related to values of FEV(1) and FEV(1)/FVC ratio, both in central and in peripheral airways. These data suggest that reduced apoptosis of CD8+ T-lymphocytes may be an important mechanism that contributes to the accumulation of these cells in the airway submucosa in smokers with mild/moderate COPD.
Collapse
Affiliation(s)
- Liboria Siena
- Istituto di Biomedicina e Immunologia Molecolare, Consiglio Nazionale delle Ricerche, Via Ugo La Malfa 153, Palermo, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Hardie DL, Baldwin MJ, Naylor A, Haworth OJ, Hou TZ, Lax S, Curnow SJ, Willcox N, MacFadyen J, Isacke CM, Buckley CD. The stromal cell antigen CD248 (endosialin) is expressed on naive CD8+ human T cells and regulates proliferation. Immunology 2011; 133:288-95. [PMID: 21466550 DOI: 10.1111/j.1365-2567.2011.03437.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
CD248 (endosialin) is a transmembrane glycoprotein that is dynamically expressed on pericytes and fibroblasts during tissue development, tumour neovascularization and inflammation. Its role in tissue remodelling is associated with increased stromal cell proliferation and migration. We show that CD248 is also uniquely expressed by human, but not mouse (C57BL/6), CD8(+) naive T cells. CD248 is found only on CD8(+) CCR7(+) CD11a(low) naive T cells and on CD8 single-positive T cells in the thymus. Transfection of the CD248 negative T-cell line MOLT-4 with CD248 cDNA surprisingly reduced cell proliferation. Knock-down of CD248 on naive CD8 T cells increased cell proliferation. These data demonstrate opposing functions for CD248 on haematopoietic (CD8(+)) versus stromal cells and suggests that CD248 helps to maintain naive CD8(+) human T cells in a quiescent state.
Collapse
Affiliation(s)
- Debbie L Hardie
- Rheumatology Research Group, MRC Centre for Immune Regulation, Institute for Biomedical Research, University of Birmingham, Edgbaston, UK
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Weitz M, Kiessling C, Friedrich M, Prösch S, Höflich C, Kern F, Volk HD, Sterry W, Asadullah K, Döcke WD. Persistent CMV infection correlates with disease activity and dominates the phenotype of peripheral CD8+ T cells in psoriasis. Exp Dermatol 2011; 20:561-7. [DOI: 10.1111/j.1600-0625.2011.01250.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
38
|
Libri V, Azevedo RI, Jackson SE, Di Mitri D, Lachmann R, Fuhrmann S, Vukmanovic-Stejic M, Yong K, Battistini L, Kern F, Soares MVD, Akbar AN. Cytomegalovirus infection induces the accumulation of short-lived, multifunctional CD4+CD45RA+CD27+ T cells: the potential involvement of interleukin-7 in this process. Immunology 2011; 132:326-39. [PMID: 21214539 DOI: 10.1111/j.1365-2567.2010.03386.x] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The relative roles that ageing and lifelong cytomegalovirus (CMV) infection have in shaping naive and memory CD4+ T-cell repertoires in healthy older people is unclear. Using multiple linear regression analysis we found that age itself is a stronger predictor than CMV seropositivity for the decrease in CD45RA+ CD27+ CD4+ T cells over time. In contrast, the increase in CD45RA⁻ CD27⁻ and CD45RA+ CD27⁻ CD4+ T cells is almost exclusively the result of CMV seropositivity, with age alone having no significant effect. Furthermore, the majority of the CD45RA⁻ CD27⁻ and CD45RA+ CD27⁻ CD4+ T cells in CMV-seropositive donors are specific for this virus. CD45RA+ CD27⁻ CD4+ T cells have significantly reduced CD28, interleukin-7 receptor α (IL-7Rα) and Bcl-2 expression, Akt (ser473) phosphorylation and reduced ability to survive after T-cell receptor activation compared with the other T-cell subsets in the same donors. Despite this, the CD45RA+ CD27⁻ subset is as multifunctional as the CD45RA⁻ D27+ and CD45RA⁻ CD27⁻ CD4+ T-cell subsets, indicating that they are not an exhausted population. In addition, CD45RA+ CD27⁻ CD4+ T cells have cytotoxic potential as they express high levels of granzyme B and perforin. CD4+ memory T cells re-expressing CD45RA can be generated from the CD45RA⁻ CD27+ population by the addition of IL-7 and during this process these cells down-regulated expression of IL-7R and Bcl-2 and so resemble their counterparts in vivo. Finally we showed that the proportion of CD45RA+ CD27⁻ CD4+ T cells of multiple specificities was significantly higher in the bone marrow than the blood of the same individuals, suggesting that this may be a site where these cells are generated.
Collapse
Affiliation(s)
- Valentina Libri
- Division of Infection and Immunity, University College London, London, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Human cytomegalovirus immunity and immune evasion. Virus Res 2010; 157:151-60. [PMID: 21056604 DOI: 10.1016/j.virusres.2010.10.031] [Citation(s) in RCA: 200] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Revised: 10/27/2010] [Accepted: 10/28/2010] [Indexed: 02/02/2023]
Abstract
Human cytomegalovirus (HCMV) infection induces both innate immune responses including Natural Killer cells as well as adaptive humoral and cell mediated (CD4+ helper, CD8+ cytotoxic and γδ T cell) responses which lead to the resolution of acute primary infection. Despite such a robust primary immune response, HCMV is still able to establish latency. Long term memory T cell responses are maintained at high frequency and are thought to prevent clinical disease following periodic reactivation of the virus. As such, a balance is established between the immune response and viral reactivation. Loss of this balance in the immunocompromised host can lead to unchecked viral replication following reactivation of latent virus, with consequent disease and mortality. HCMV encodes multiple immune evasion mechanisms that target both the innate and acquired immune system. This article describes the current understanding of Natural killer cell, antibody and T cell mediated immune responses and the mechanisms that the virus utilizes to subvert these responses.
Collapse
|
40
|
Jafari N, Broer L, Hoppenbrouwers IA, van Duijn CM, Hintzen RQ. Infectious mononucleosis-linked HLA class I single nucleotide polymorphism is associated with multiple sclerosis. Mult Scler 2010; 16:1303-7. [DOI: 10.1177/1352458510376778] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background: Multiple sclerosis is a presumed autoimmune disease associated with genetic and environmental risk factors such as infectious mononucleosis. Recent research has shown infectious mononucleosis to be associated with a specific HLA class I polymorphism. Objectives: Our aim was to test if the infectious mononucleosis-linked HLA class I single nucleotide polymorphism (rs6457110) is also associated with multiple sclerosis. Methods: Genotyping of the HLA-A single nucleotide polymorphism rs6457110 using TaqMan was performed in 591 multiple sclerosis cases and 600 controls. The association of multiple sclerosis with the HLA-A single nucleotide polymorphism was tested using logistic regression adjusted for age, sex and HLA-DRB1*1501. Results: HLA-A minor allele (A) is associated with multiple sclerosis (OR = 0.68; p = 4.08 × 10 -5). After stratification for HLA-DRB1*1501 risk allele (T) carrier we showed a significant OR of 0.70 ( p = 0.003) for HLA-A. Conclusions: HLA class I single nucleotide polymorphism rs6457110 is associated with infectious mononucleosis and multiple sclerosis, independent of the major class II allele, supporting the hypothesis that shared genetics may contribute to the association between infectious mononucleosis and multiple sclerosis.
Collapse
Affiliation(s)
- Naghmeh Jafari
- Department of Neurology, ErasMS Centre, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Linda Broer
- Department of Epidemiology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Ilse A Hoppenbrouwers
- Department of Neurology, ErasMS Centre, Erasmus Medical Centre, Rotterdam, The Netherlands
| | | | - Rogier Q Hintzen
- Department of Neurology, ErasMS Centre, Erasmus Medical Centre, Rotterdam, The Netherlands,
| |
Collapse
|
41
|
Lima K, Abrahamsen TG, Foelling I, Natvig S, Ryder LP, Olaussen RW. Low thymic output in the 22q11.2 deletion syndrome measured by CCR9+CD45RA+ T cell counts and T cell receptor rearrangement excision circles. Clin Exp Immunol 2010; 161:98-107. [PMID: 20491792 DOI: 10.1111/j.1365-2249.2010.04152.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Thymic hypoplasia is a frequent feature of the 22q11.2 deletion syndrome, but we know little about patients' age-related thymic output and long-term consequences for their immune system. We measured the expression of T cell receptor rearrangement excision circles (TREC) and used flow cytometry for direct subtyping of recent thymic emigrant (RTE)-related T cells in 43 patients (aged 1-54 years; median 9 years) from all over Norway and in age-matched healthy controls. Thymic volumes were estimated by ultrasound in patients. TREC levels correlated well with RTE-related T cells defined by co-expression of CD3, CD45RA and CCR9 (r=0.84) as well as with the CD4+ and CD8+ T cell subtypes. RTE-related T cell counts also paralleled age-related TREC reductions. CD45RA+ T cells correlated well with absolute counts of CD4+ (r=0.87) and CD8+ (r=0.75) RTE-related T cells. Apart from CD45RA- T cells, all T cell subsets were lower in patients than in controls. Thymic volumes correlated better with RTE-related cells (r=0.46) than with TREC levels (r=0.38). RTE-related T cells and TREC levels also correlated well (r=0.88) in patients without an identifiable thymus. Production of RTEs is impaired in patients with a 22q11.2 deletion, and CCR9 appears to be a good marker for RTE-related T cells.
Collapse
Affiliation(s)
- K Lima
- Section of Endocrinology, Faculty Division Akershus University Hospital, University of Oslo, Oslo, Norway.
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
Chemokines direct leukocyte trafficking and positioning within tissues, thus playing critical roles in regulating immune responses and inflammation. The chemokine system is complex, involving interactions between multiple chemokines and their receptors that operate in combinatorial cascades with adhesion molecules. The involvement of multiple chemokines and chemokine receptors in these processes brings flexibility and specificity to recruitment. The hepatic vascular bed is a unique low-flow environment through which leukocytes are recruited to the liver during homeostatic immune surveillance and in response to infection or injury. The rate of leukocyte recruitment and the nature of cells recruited through the sinusoids in response to inflammatory signals will shape the severity of disease. At one end of the spectrum, fulminant liver failure results from a rapid recruitment of leukocytes that leads to hepatocyte destruction and liver failure; at the other end, diseases such as chronic hepatitis C infection may progress over many years from hepatitis to fibrosis and cirrhosis. Chronic hepatitis is characterized by a T lymphocyte-rich infiltrate and the nature and outcome of hepatitis will depend on the T cell subsets recruited, their activation and function within the liver. Different subsets of effector T cells have been described based on their secretion of cytokines and specific functions. These include Th1 and Th2 cells, and more recently Th17 and Th9 cells, which are associated with different types of immune response and which express distinct patterns of chemokine receptors that promote their recruitment under particular conditions. The effector function of these cells is balanced by the recruitment of regulatory T cells that are able to suppress antigen-specific effectors to allow resolution of immune responses and restoration of immune homeostasis. Understanding the signals that are responsible for recruiting different lymphocyte subsets to the liver will elucidate disease pathogenesis and open up new therapeutic approaches to modulate recruitment in favor of resolution rather than injury.
Collapse
Affiliation(s)
| | | | - David H. Adams
- *Prof. David H. Adams, MD, FRCP, FmedSci, 5th Floor, Institute of Biomedical Research, University of Birmingham Medical School, Wolfson Drive, Edgbaston, Birmingham B15 2TT (UK), Tel. +44 121 415 8702, Fax +44 121 415 8701, E-Mail
| |
Collapse
|
43
|
Makedonas G, Hutnick N, Haney D, Amick AC, Gardner J, Cosma G, Hersperger AR, Dolfi D, Wherry EJ, Ferrari G, Betts MR. Perforin and IL-2 upregulation define qualitative differences among highly functional virus-specific human CD8 T cells. PLoS Pathog 2010; 6:e1000798. [PMID: 20221423 PMCID: PMC2832688 DOI: 10.1371/journal.ppat.1000798] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2009] [Accepted: 01/28/2010] [Indexed: 01/12/2023] Open
Abstract
The prevailing paradigm of T lymphocyte control of viral replication is that the protective capacity of virus-specific CD8+ T cells is directly proportional to the number of functions they can perform, with IL-2 production capacity considered critical. Having recently defined rapid perforin upregulation as a novel effector function of antigen-specific CD8+ T cells, here we sought to determine whether new perforin production is a component of polyfunctional CD8+ T cell responses that contributes to the control of several human viral infections: cytomegalovirus (CMV), Epstein-Barr virus (EBV), influenza (flu), and adenovirus (Ad). We stimulated normal human donor PBMC with synthetic peptides whose amino acid sequences correspond to defined CTL epitopes in the aforementioned viruses, and then used polychromatic flow cytometry to measure the functional capacity and the phenotype of the responding CD8+ T cells. While EBV and flu-specific CD8+ T cells rarely upregulate perforin, CMV-specific cells often do and Ad stimulates an exceptionally strong perforin response. The differential propensity of CD8+ T cells to produce either IL-2 or perforin is in part related to levels of CD28 and the transcription factor T-bet, as CD8+ T cells that rapidly upregulate perforin harbor high levels of T-bet and those producing IL-2 express high amounts of CD28. Thus, “polyfunctional” profiling of antigen-specific CD8+ T cells must not be limited to simply the number of functions the cell can perform, or one particular memory phenotype, but should actually define which combinations of memory markers and functions are relevant in each pathogenic context. Although CD8+ T cells are thought to be largely responsible for the control of viral infections, exactly how they mediate protection is uncertain. One approach to assessing their protective capacity is to measure several of their functions simultaneously. Generally, it is believed the more functions a cell can perform, the better its potential to control viral replication. A multi-functional response including interleukin-2 (IL-2) production is currently valued as the key correlate of protection. We recently characterized a novel CD8+ T cell function: rapid perforin upregulation, which serves to contribute to and sustain the killing of virally infected host cells. In this study, we show that new perforin is abundant during adenovirus and cytomegalovirus infections, but scarcely detected in the context of influenza and Epstein-Barr virus. Importantly, perforin and IL-2 are rarely co-expressed. The significance of this relationship is that we can no longer assume the more functions a CD8+ T cell performs in response to a virus the better. Thus, when considering vaccine design, no single functional profile will likely be protective across all pathogens. Rather, vaccine-induced T cell responses may need to be “pathogen-specific”, as different T cell functional responses will be important for controlling different viral infections.
Collapse
Affiliation(s)
- George Makedonas
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Natalie Hutnick
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Danielle Haney
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Alexandra C. Amick
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jay Gardner
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Gabriela Cosma
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Adam R. Hersperger
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Douglas Dolfi
- The Wistar Institute, Philadelphia, Pennsylvania, United States of America
| | - E. John Wherry
- The Wistar Institute, Philadelphia, Pennsylvania, United States of America
| | - Guido Ferrari
- Department of Surgical Sciences, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Michael R. Betts
- Department of Microbiology, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
44
|
Henson SM, Akbar AN. Memory T-Cell Homeostasis and Senescence during Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 684:189-97. [DOI: 10.1007/978-1-4419-6451-9_15] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
45
|
Henson SM, Akbar AN. KLRG1--more than a marker for T cell senescence. AGE (DORDRECHT, NETHERLANDS) 2009; 31:285-91. [PMID: 19479342 PMCID: PMC2813054 DOI: 10.1007/s11357-009-9100-9] [Citation(s) in RCA: 150] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2009] [Accepted: 05/14/2009] [Indexed: 05/17/2023]
Abstract
The co-inhibitory receptor killer-cell lectin like receptor G1 (KLRG1) is expressed on NK cells and antigen-experienced T cells and has been postulated to be a marker of senescence. Whilst KLRG1 has frequently been used as a marker of cellular differentiation, data are emerging indicating that KLRG1 plays an inhibitory role. In this review we examine evidence highlighting this view of KLRG1 with emphasis on the functional defects that arise during T cell differentiation with age that may, in part, be actively maintained by inhibitory receptor signalling.
Collapse
Affiliation(s)
- Sian M Henson
- Division of Infection and Immunity, Department of Immunology, University College London, 46 Cleveland Street, London, UK.
| | | |
Collapse
|
46
|
Oo YH, Adams DH. The role of chemokines in the recruitment of lymphocytes to the liver. J Autoimmun 2009; 34:45-54. [PMID: 19744827 DOI: 10.1016/j.jaut.2009.07.011] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2009] [Accepted: 07/29/2009] [Indexed: 12/13/2022]
Abstract
Chemokines direct leukocyte trafficking and positioning within tissues. They thus play critical roles in regulating immune responses and inflammation. The chemokine system is complex involving interactions between multiple chemokines and their receptors that operate in combinatorial cascades with adhesion molecules. The involvement of multiple chemokines and chemokine receptors in these processes brings flexibility and specificity to recruitment. The hepatic vascular bed is a unique low flow environment through which leukocyte are recruited to the liver during homeostatic immune surveillance and in response to infection or injury. The rate of leukocyte recruitment and the nature of cells recruited through the sinusoids in response to inflammatory signals will shape the severity of disease. At one end of the spectrum fulminant liver failure results from a rapid recruitment of leukocytes that leads to hepatocyte destruction and liver failure at the other diseases such as chronic hepatitis C infection may progress over many years from hepatitis to fibrosis and cirrhosis. Chronic hepatitis is charactezised by a T lymphocyte rich infiltrate and the nature and outcome of hepatitis will depend on the T cell subsets recruited, their activation and function within the liver. Different subsets of effector T cells have been described based on their secretion of cytokines and specific functions. These include Th1 and Th2 cells and more recently Th17 and Th9 cells which are associated with different types of immune response and which express distinct patterns of chemokine receptors that promote their recruitment under particular conditions. The effector function of these cells is balanced by the recruitment of regulatory T cells that are able to suppress antigen-specific effectors to allow resolution of immune responses and restoration of immune homeostasis. Understanding the signals that are responsible for recruiting different lymphocyte subsets to the liver will elucidate disease pathogenesis and open up new therapeutic approaches to modulate recruitment in favour of resolution rather than injury.
Collapse
Affiliation(s)
- Ye H Oo
- Centre for Liver Research, 5th Floor, Institute of Biomedical Research, University of Birmingham, Wolfson Drive, Edgbaston, Birmingham B15 2TT, UK
| | | |
Collapse
|
47
|
Campbell JP, Riddell NE, Burns VE, Turner M, van Zanten JJCSV, Drayson MT, Bosch JA. Acute exercise mobilises CD8+ T lymphocytes exhibiting an effector-memory phenotype. Brain Behav Immun 2009; 23:767-75. [PMID: 19254756 DOI: 10.1016/j.bbi.2009.02.011] [Citation(s) in RCA: 148] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2008] [Revised: 02/17/2009] [Accepted: 02/19/2009] [Indexed: 12/24/2022] Open
Abstract
An acute bout of exercise evokes mobilisation of lymphocytes into the bloodstream, which can be largely attributed to increases in CD8+ T lymphocytes (CD8TLs) and natural killer (NK) cells. Evidence further suggests that, even within these lymphocyte subsets, there is preferential mobilisation of cells that share certain functional and phenotypic characteristics, such as high cytotoxicity, low proliferative ability, and high tissue-migrating potential. These features are characteristic of effector-memory CD8TL subsets. The current study therefore investigated the effect of exercise on these newly-identified subsets. Thirteen healthy and physically active males (mean+/-SD: age 20.9+/-1.5 yr) attended three sessions: a control session (no exercise); cycling at 35% Watt(max) (low intensity exercise); and 85% Watt(max) (high intensity exercise). Each bout lasted 20 min. Blood samples were obtained before exercise, during the final min of exercise, and +15, and +60 min post-exercise. CD8TLs were classified into naïve, central memory (CM), effector-memory (EM), and CD45RA+ effector-memory (RAEM) using combinations of the cell surface markers CCR7, CD27, CD62L, CD57, and CD45RA. In parallel, the phenotypically distinct CD56(bright) 'regulatory' and CD56(dim) 'cytotoxic' NK subsets were quantified. The results show a strong differential mobilisation of CD8TL subsets (RAEM>EM>CM>naïve); during high intensity exercise the greatest increase was observed for RAEM CD8Tls (+450%) and the smallest for naïve cells (+84%). Similarly, CD56(dim) NK cells (+995%) were mobilised to a greater extent than CD56(bright) (+153%) NK cells. In conclusion, memory CD8TL that exhibit a high effector and tissue-migrating potential are preferentially mobilised during exercise. This finding unifies a range of independent observations regarding exercise-induced phenotypic and functional changes in circulating lymphocytes. The selective mobilisation of cytotoxic tissue-migrating subsets, both within the NK and CD8TL population, may enhance immune-surveillance during exercise.
Collapse
Affiliation(s)
- John P Campbell
- Behavioural Medicine Group, School of Sport and Exercise Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | | | | | | | | | | | | |
Collapse
|
48
|
Antiretroviral therapy restores diversity in the T-cell receptor Vbeta repertoire of CD4 T-cell subpopulations among human immunodeficiency virus type 1-infected children and adolescents. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2009; 16:1293-301. [PMID: 19605599 DOI: 10.1128/cvi.00074-09] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Human immunodeficiency virus (HIV) type 1 infection perturbs the T-cell receptor (TCR) Vbeta repertoire. The TCR CDR3 length diversity of individual Vbeta families was examined within CD45RA and CD45RO CD4 T cells to assess the impact of the virus on clonality throughout CD4 T-cell activation and differentiation. A cross-sectional and longitudinal cohort study of 13 HIV-infected and 8 age-matched healthy children and adolescents examined the Vbeta CDR3 length profiles within CD4 T-cell subsets by the use of spectratyping. HIV-infected subjects demonstrated higher numbers of perturbations in CD4 CD45RA T cells (5.8 +/- 4.9 Vbeta families) than healthy individuals (1.6 +/- 1.8 Vbeta families) (P = 0.04). Surprisingly, CD4 CD45RO central memory T cells from infected subjects showed no increased perturbations compared to the perturbations for the same cells from healthy subjects (2.9 +/- 3.1 and 1.1 +/- 1.8 Vbeta families, respectively; P = 0.11). CD4 CD45RA TCR perturbations were higher among infected subjects with >25% CD4 cells than healthy subjects (mean number of perturbed Vbeta families, 6.6 +/- 5.4; P = 0.04). No correlations between perturbations in CD4 subsets and pretherapy age or viral load were evident. In contrast to CD8 T cells, HIV induces TCR disruptions within CD45RA but not CD45RO CD4 T cells. Therapy-induced viral suppression resulted in increases in thymic output and the normalization of the diversity of TCR within CD45RA CD4 T cells after 2 months of treatment. Perturbations occur prior to CD4 T-cell attrition and normalize with effective antiretroviral therapy. The impact of HIV on the diversity of TCR within naïve, central memory, and effector memory CD4 T cells is distinctly different from that in CD8 T cells.
Collapse
|
49
|
Belmonte L, Parodi C, Bastón M, Coraglia A, Felippo M, Baré P, Malbrán A, Ruibal-Ares B, de E de Bracco MM. Increased lymphocyte viability after non-stimulated peripheral blood mononuclear cell (PBMC) culture in patients with X-linked lymphoproliferative disease (XLP). Clin Immunol 2009; 133:86-94. [PMID: 19576857 DOI: 10.1016/j.clim.2009.05.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2009] [Revised: 05/19/2009] [Accepted: 05/27/2009] [Indexed: 10/20/2022]
Abstract
Survival of lymphocytes after prolonged culture was studied in two asymptomatic XLP patients. Viability of XLP PBMC after 30 days of non-stimulated culture was higher than that of normal controls (N), mainly due to the persistence of CD8 memory lymphocytes. IFNgamma high CD8 T lymphocytes remained higher in XLP than in N after 30 days. The number of perforin+ CD8 lymphocytes was markedly reduced after 30 days in XLP and in N. Increased viability was not related to CD127, PD-1, CD27, or CD62L expression. Concerning B lymphocytes, memory CD27+ CD19+ cells prevailed over CD27- cells after 30 days in both XLP and N, with far more surviving cells in XLP. In N, few CD19+ B lymphocytes were viable after prolonged culture. In XLP, these cells were also IgD+, IgM+ and EBNA2+. These results demonstrate that IFNgamma-positive memory CD8 T cells persist in XLP after prolonged culture in association with a subset of viable memory CD27+ B cells expressing latent EBV antigens. The survival advantage of XLP cells might be related to increased frequency of extranodal lymphoma in XLP patients.
Collapse
Affiliation(s)
- Liliana Belmonte
- Instituto de Investigaciones Hematológicas, Academia Nacional de Medicina, 1425 Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Bruns H, Meinken C, Schauenberg P, Härter G, Kern P, Modlin RL, Antoni C, Stenger S. Anti-TNF immunotherapy reduces CD8+ T cell-mediated antimicrobial activity against Mycobacterium tuberculosis in humans. J Clin Invest 2009; 119:1167-77. [PMID: 19381021 DOI: 10.1172/jci38482] [Citation(s) in RCA: 230] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2009] [Accepted: 03/25/2009] [Indexed: 12/22/2022] Open
Abstract
The incidence of tuberculosis is increased during treatment of autoimmune diseases with anti-TNF antibodies. This is a significant clinical complication, but also provides a unique model to study immune mechanisms in human tuberculosis. Given the key role for cell-mediated immunity in host defense against Mycobacterium tuberculosis, we hypothesized that anti-TNF treatment impairs T cell-directed antimicrobial activity. Anti-TNF therapy reduced the expression in lymphocytes of perforin and granulysin, 2 components of the T cell-mediated antimicrobial response to intracellular pathogens. Specifically, M. tuberculosis-reactive CD8+CCR7-CD45RA+ effector memory T cells (TEMRA cells) expressed the highest levels of granulysin, lysed M. tuberculosis, and infected macrophages and mediated an antimicrobial activity against intracellular M. tuberculosis. Furthermore, TEMRA cells expressed cell surface TNF and bound the anti-TNF therapeutic infliximab in vitro, making them susceptible to complement-mediated lysis. Immune therapy with anti-TNF was associated with reduced numbers of CD8+ TEMRA cells and decreased antimicrobial activity against M. tuberculosis, which could be rescued by the addition of CD8+ TEMRA cells. These results suggest that anti-TNF therapy triggers a reduction of CD8+ TEMRA cells with antimicrobial activity against M. tuberculosis, providing insight into the mechanism whereby key effector T cell subsets contribute to host defense against tuberculosis.
Collapse
Affiliation(s)
- Heiko Bruns
- Institute for Medical Microbiology and Hygiene, University Hospital of Ulm, Germany
| | | | | | | | | | | | | | | |
Collapse
|