1
|
Wohlfarth J, Kosnopfel C, Faber D, Berthold M, Siedel C, Bernhardt M, Schlosser A, Aprati T, Liu D, Schrama D, Houben R, Schadendorf D, Goebeler M, Meierjohann S, Schilling B. Loss of p14 diminishes immunogenicity in melanoma via non-canonical Wnt signaling by reducing the peptide surface density. Mol Oncol 2024; 18:2449-2470. [PMID: 38807304 PMCID: PMC11459041 DOI: 10.1002/1878-0261.13660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/04/2024] [Accepted: 04/26/2024] [Indexed: 05/30/2024] Open
Abstract
Immunotherapy has achieved tremendous success in melanoma. However, only around 50% of advanced melanoma patients benefit from immunotherapy. Cyclin-dependent kinase inhibitor 2A (CDKN2A), encoding the two tumor-suppressor proteins p14ARF and p16INK4a, belongs to the most frequently inactivated gene loci in melanoma and leads to decreased T cell infiltration. While the role of p16INK4a has been extensively investigated, knowledge about p14ARF in melanoma is scarce. In this study, we elucidate the impact of reduced p14ARF expression on melanoma immunogenicity. Knockdown of p14ARF in melanoma cell lines diminished their recognition and killing by melanoma differentiation antigen (MDA)-specific T cells. Resistance was caused by a reduction of the peptide surface density of presented MDAs. Immunopeptidomic analyses revealed that antigen presentation via human leukocyte antigen class I (HLA-I) molecules was enhanced upon p14ARF downregulation in general, but absolute and relative expression of cognate peptides was decreased. However, this phenotype is associated with a favorable outcome for melanoma patients. Limiting Wnt5a signaling reverted this phenotype, suggesting an involvement of non-canonical Wnt signaling. Taken together, our data indicate a new mechanism limiting MDA-specific T cell responses by decreasing both absolute and relative MDA-peptide presentation in melanoma.
Collapse
Affiliation(s)
- Jonas Wohlfarth
- Department of Dermatology, Venereology and AllergologyUniversity Hospital WürzburgGermany
| | - Corinna Kosnopfel
- Department of Dermatology, Venereology and AllergologyUniversity Hospital WürzburgGermany
| | - Dominic Faber
- Department of Dermatology, Venereology and AllergologyUniversity Hospital WürzburgGermany
| | - Marion Berthold
- Department of Dermatology, Venereology and AllergologyUniversity Hospital WürzburgGermany
| | - Claudia Siedel
- Department of Dermatology, Venereology and AllergologyUniversity Hospital WürzburgGermany
| | - Melissa Bernhardt
- Rudolf‐Virchow‐Centre for Integrative and Translational BioimagingUniversity of WürzburgGermany
| | - Andreas Schlosser
- Rudolf‐Virchow‐Centre for Integrative and Translational BioimagingUniversity of WürzburgGermany
| | - Tyler Aprati
- Dana‐Farber Cancer InstituteBostonMAUSA
- Harvard Medical SchoolCambridgeMAUSA
- Broad Institute of Harvard and MITCambridgeMAUSA
| | - David Liu
- Dana‐Farber Cancer InstituteBostonMAUSA
- Harvard Medical SchoolCambridgeMAUSA
- Broad Institute of Harvard and MITCambridgeMAUSA
| | - David Schrama
- Department of Dermatology, Venereology and AllergologyUniversity Hospital WürzburgGermany
| | - Roland Houben
- Department of Dermatology, Venereology and AllergologyUniversity Hospital WürzburgGermany
| | - Dirk Schadendorf
- Department of Dermatology, Comprehensive Cancer Center (Westdeutsches Tumorzentrum)German Cancer Consortium (DKTK, partner site Essen) and University Hospital EssenGermany
| | - Matthias Goebeler
- Department of Dermatology, Venereology and AllergologyUniversity Hospital WürzburgGermany
| | | | - Bastian Schilling
- Department of Dermatology, Venereology and AllergologyUniversity Hospital WürzburgGermany
| |
Collapse
|
2
|
Linette GP, Bear AS, Carreno BM. Facts and Hopes in Immunotherapy Strategies Targeting Antigens Derived from KRAS Mutations. Clin Cancer Res 2024; 30:2017-2024. [PMID: 38266167 PMCID: PMC11094419 DOI: 10.1158/1078-0432.ccr-23-1212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/20/2023] [Accepted: 01/03/2024] [Indexed: 01/26/2024]
Abstract
In this commentary, we advance the notion that mutant KRAS (mKRAS) is an ideal tumor neoantigen that is amenable for targeting by the adaptive immune system. Recent progress highlights key advances on various fronts that validate mKRAS as a molecular target and support further pursuit as an immunological target. Because mKRAS is an intracellular membrane localized protein and not normally expressed on the cell surface, we surmise that proteasome degradation will generate short peptides that bind to HLA class I (HLA-I) molecules in the endoplasmic reticulum for transport through the Golgi for display on the cell surface. T-cell receptors (TCR)αβ and antibodies have been isolated that specifically recognize mKRAS encoded epitope(s) or haptenated-mKRAS peptides in the context of HLA-I on tumor cells. Case reports using adoptive T-cell therapy provide proof of principle that KRAS G12D can be successfully targeted by the immune system in patients with cancer. Among the challenges facing investigators is the requirement of precision medicine to identify and match patients to available mKRAS peptide/HLA therapeutics and to increase the population coverage by targeting additional mKRAS epitopes. Ultimately, we envision mKRAS-directed immunotherapy as an effective treatment option for selected patients that will complement and perhaps synergize with small-molecule mKRAS inhibitors and targeted mKRAS degraders.
Collapse
Affiliation(s)
- Gerald P. Linette
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Adham S. Bear
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Beatriz M. Carreno
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
3
|
Heidarian M, Jensen IJ, Kannan SK, Pewe LL, Hassert M, Park S, Xue HH, Harty JT, Badovinac VP. Sublethal whole-body irradiation induces permanent loss and dysfunction in pathogen-specific circulating memory CD8 T cell populations. Proc Natl Acad Sci U S A 2023; 120:e2302785120. [PMID: 37364124 PMCID: PMC10318958 DOI: 10.1073/pnas.2302785120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/31/2023] [Indexed: 06/28/2023] Open
Abstract
The increasing use of nuclear energy sources inevitably raises the risk of accidental or deliberate radiation exposure and associated immune dysfunction. However, the extent to which radiation exposure impacts memory CD8 T cells, potent mediators of immunity to recurring intracellular infections and malignancies, remains understudied. Using P14 CD8 T cell chimeric mice (P14 chimeras) with an lymphocytic choriomeningitis virus (LCMV) infection model, we observed that sublethal (5Gy) whole-body irradiation (WBI) induced a rapid decline in the number of naive (TN) and P14 circulating memory CD8 T cells (TCIRCM), with the former being more susceptible to radiation-induced numeric loss. While TN cell numbers rapidly recovered, as previously described, the number of P14 TCIRCM cells remained low at least 9 mo after radiation exposure. Additionally, the remaining P14 TCIRCM in irradiated hosts exhibited an inefficient transition to a central memory (CD62L+) phenotype compared to nonirradiated P14 chimeras. WBI also resulted in long-lasting T cell intrinsic deficits in memory CD8 T cells, including diminished cytokine and chemokine production along with impaired secondary expansion upon cognate Ag reencounter. Irradiated P14 chimeras displayed significantly higher bacterial burden after challenge with Listeria monocytogenes expressing the LCMV GP33-41 epitope relative to nonirradiated controls, likely due to radiation-induced numerical and functional impairments. Taken together, our findings suggest that sublethal radiation exposure caused a long-term numerical, impaired differentiation, and functional dysregulation in preexisting TCIRCM, rendering previously protected hosts susceptible to reinfection.
Collapse
Affiliation(s)
| | - Isaac J. Jensen
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA52246
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY10032
| | - Shravan Kumar Kannan
- Department of Pathology, University of Iowa, Iowa City, IA52246
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA52246
| | - Lecia L. Pewe
- Department of Pathology, University of Iowa, Iowa City, IA52246
| | - Mariah Hassert
- Department of Pathology, University of Iowa, Iowa City, IA52246
| | - SungRye Park
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ07110
| | - Hai-Hui Xue
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ07110
| | - John T. Harty
- Department of Pathology, University of Iowa, Iowa City, IA52246
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA52246
| | - Vladimir P. Badovinac
- Department of Pathology, University of Iowa, Iowa City, IA52246
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA52246
| |
Collapse
|
4
|
Landscape of Exhausted Virus-Specific CD8 T Cells in Chronic LCMV Infection. Cell Rep 2021; 32:108078. [PMID: 32846135 DOI: 10.1016/j.celrep.2020.108078] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 01/31/2020] [Accepted: 08/05/2020] [Indexed: 01/14/2023] Open
Abstract
A hallmark of chronic infections is the presence of exhausted CD8 T cells, characterized by a distinct transcriptional program compared with functional effector or memory cells, co-expression of multiple inhibitory receptors, and impaired effector function, mainly driven by recurrent T cell receptor engagement. In the context of chronic lymphocytic choriomeningitis virus (LCMV) infection in mice, most studies focused on studying splenic virus-specific CD8 T cells. Here, we provide a detailed characterization of exhausted CD8 T cells isolated from six different tissues during established LCMV infection, using single-cell RNA sequencing. Our data reveal that exhausted cells are heterogeneous, adopt organ-specific transcriptomic profiles, and can be divided into five main functional subpopulations: advanced exhaustion, effector-like, intermediate, proliferating, or memory-like. Adoptive transfer experiments showed that these phenotypes are plastic, suggesting that the tissue microenvironment has a major impact in shaping the phenotype and function of virus-specific CD8 T cells during chronic infection.
Collapse
|
5
|
Chaoul N, Dadaglio G. Antigen-Specific In Vivo Killing Assay. Methods Mol Biol 2021; 2325:55-64. [PMID: 34053050 DOI: 10.1007/978-1-0716-1507-2_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The in vivo killing assay allows the quantification of the antigen-specific killing capacity of Cytotoxic CD8+ T Lymphocytes (CTLs) in mice. CTLs are indeed known for the lysis of cells expressing foreign or modified antigen peptides on their MHC class I molecules. Here we describe the detailed protocol used for the in vivo specific lysis of cells expressing the H-2 Kb immunodominant CD8+ T-cell epitope of the OVA protein: an 8 amino acid peptide corresponding to the 257-264 region of OVA (SIINFEKL).
Collapse
Affiliation(s)
- Nada Chaoul
- Department of Emergency and Organ Transplant, School and Chair of Allergology and Clinical Immunology, University of Bari-Aldo Moro, Bari, Italy.
| | - Gilles Dadaglio
- Department of Immunology, Immunobiology Infection Unit, Institut Pasteur, INSERM U1221, Paris, France
| |
Collapse
|
6
|
Exhausted CD8 + T cells exhibit low and strongly inhibited TCR signaling during chronic LCMV infection. Nat Commun 2020; 11:4454. [PMID: 32901001 PMCID: PMC7479152 DOI: 10.1038/s41467-020-18256-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 08/12/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic viral infections are often associated with impaired CD8+ T cell function, referred to as exhaustion. Although the molecular and cellular circuits involved in CD8+ T cell exhaustion are well defined, with sustained presence of antigen being one important parameter, how much T cell receptor (TCR) signaling is actually ongoing in vivo during established chronic infection is unclear. Here, we characterize the in vivo TCR signaling of virus-specific exhausted CD8+ T cells in a mouse model, leveraging TCR signaling reporter mice in combination with transcriptomics. In vivo signaling in exhausted cells is low, in contrast to their in vitro signaling potential, and despite antigen being abundantly present. Both checkpoint blockade and adoptive transfer of naïve target cells increase TCR signaling, demonstrating that engagement of co-inhibitory receptors curtails CD8+ T cell signaling and function in vivo. Excess antigenic exposure, such as in cancers or chronic viral infection, can lead to T cell exhaustion. Here the authors show that despite high exposure to antigen in the context of chronic LCMV infection in mice, exhausted CD8+ T cells have low levels of TCR signalling that can be reactivated by PD-L1 blockade.
Collapse
|
7
|
Garcia V, Bonhoeffer S, Fu F. Cancer-induced immunosuppression can enable effectiveness of immunotherapy through bistability generation: A mathematical and computational examination. J Theor Biol 2020; 492:110185. [PMID: 32035826 PMCID: PMC7079339 DOI: 10.1016/j.jtbi.2020.110185] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 01/09/2020] [Accepted: 02/03/2020] [Indexed: 12/22/2022]
Abstract
The presence of an immunological barrier in cancer- immune system interaction (CISI) is consistent with the bistability patterns in that system. In CISI models, bistability patterns are consistent with immunosuppressive effects dominating immunoproliferative effects. Bistability could be harnessed to devise effective combination immunotherapy approaches.
Cancer immunotherapies rely on how interactions between cancer and immune system cells are constituted. The more essential to the emergence of the dynamical behavior of cancer growth these interactions are, the more effectively they may be used as mechanisms for interventions. Mathematical modeling can help unearth such connections, and help explain how they shape the dynamics of cancer growth. Here, we explored whether there exist simple, consistent properties of cancer-immune system interaction (CISI) models that might be harnessed to devise effective immunotherapy approaches. We did this for a family of three related models of increasing complexity. To this end, we developed a base model of CISI, which captures some essential features of the more complex models built on it. We find that the base model and its derivates can plausibly reproduce biological behavior that is consistent with the notion of an immunological barrier. This behavior is also in accord with situations in which the suppressive effects exerted by cancer cells on immune cells dominate their proliferative effects. Under these circumstances, the model family may display a pattern of bistability, where two distinct, stable states (a cancer-free, and a full-grown cancer state) are possible. Increasing the effectiveness of immune-caused cancer cell killing may remove the basis for bistability, and abruptly tip the dynamics of the system into a cancer-free state. Additionally, in combination with the administration of immune effector cells, modifications in cancer cell killing may be harnessed for immunotherapy without the need for resolving the bistability. We use these ideas to test immunotherapeutic interventions in silico in a stochastic version of the base model. This bistability-reliant approach to cancer interventions might offer advantages over those that comprise gradual declines in cancer cell numbers.
Collapse
Affiliation(s)
- Victor Garcia
- Institute of Applied Simulation, Zurich University of Applied Sciences, Einsiedlerstrasse 31a, 8820 Wädenswil, Switzerland; ETH Zurich, Universitätstrasse 16, 8092 Zürich, Switzerland; Institute for Social and Preventive Medicine, University of Bern, Finkenhubelweg 11, 3012 Bern, Switzerland; Department of Biology, Stanford University, 371 Serra Mall, Stanford CA 94305, USA.
| | | | - Feng Fu
- Department of Mathematics, Dartmouth College, 27 N. Main Street, 6188 Kemeny Hall, Hanover, NH 03755-3551, USA; ETH Zurich, Universitätstrasse 16, 8092 Zürich, Switzerland
| |
Collapse
|
8
|
Halle S, Halle O, Förster R. Mechanisms and Dynamics of T Cell-Mediated Cytotoxicity In Vivo. Trends Immunol 2017; 38:432-443. [PMID: 28499492 DOI: 10.1016/j.it.2017.04.002] [Citation(s) in RCA: 216] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/05/2017] [Accepted: 04/06/2017] [Indexed: 02/06/2023]
Abstract
Cytotoxic T lymphocytes (CTLs) are critical in the elimination of infected or malignant cells and are emerging as a major therapeutic target. How CTLs recognize and kill harmful cells has been characterized in vitro but little is known about these processes in the living organism. Here we review recent insights into CTL-mediated killing with an emphasis on in vivo CTL biology. Specifically, we focus on the possible rate-limiting steps determining the efficiency of CTL-mediated killing. We also highlight the need for cell-based datasets that permit the quantification of CTL dynamics, including CTL location, migration, and killing rates. A better understanding of these factors is required to predict protective CD8 T cell immunity in vivo and to design optimized vaccination protocols.
Collapse
Affiliation(s)
- Stephan Halle
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany.
| | - Olga Halle
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany.
| |
Collapse
|
9
|
Lee Y, Kim J, An J, Lee H, Kong H, Song Y, Shin E, Do SG, Lee CK, Kim K. Aloe QDM complex enhances specific cytotoxic T lymphocyte killing in vivo in metabolic disease mice. Biosci Biotechnol Biochem 2016; 81:595-603. [PMID: 27884090 DOI: 10.1080/09168451.2016.1258986] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We developed spontaneous diet-induced metabolic disease in mice by feeding them a high-fat diet for 23 weeks and administered Aloe QDM complex for 16 weeks to examine its restorative effect on immune disorders and metabolic syndrome. A series of immune functional assays indicated Aloe QDM complex enhanced lymphocyte proliferation and antigen-specific immunity as determined by the restored functions of cytotoxic T lymphocytes (CTL) and IgG production. The elevated serum TNF-α level was also regulated by Aloe QDM complex treatment, which suggested its complex therapeutic potential. As for metabolic phenotypes, oral administration of Aloe QDM complex significantly improved diabetic symptoms, including high fasting glucose levels and glucose tolerance, and distinctly alleviated lipid accumulation in adipose and hepatic tissue. The simultaneous restoration of Aloe QDM complex on metabolic syndrome and host immune dysfunction, especially on the specific CTL killing was first elucidated in our study.
Collapse
Affiliation(s)
- Youngjoo Lee
- a College of Pharmacy, Sahmyook University , Seoul , Korea
| | - Jiyeon Kim
- a College of Pharmacy, Sahmyook University , Seoul , Korea
| | - Jinho An
- a College of Pharmacy, Sahmyook University , Seoul , Korea
| | - Heetae Lee
- a College of Pharmacy, Sahmyook University , Seoul , Korea
| | - Hyunseok Kong
- a College of Pharmacy, Sahmyook University , Seoul , Korea
| | | | - Eunju Shin
- b Wellness R&D Center, Univera, Inc. , Seoul , Korea
| | - Seon-Gil Do
- b Wellness R&D Center, Univera, Inc. , Seoul , Korea
| | - Chong-Kil Lee
- c College of Pharmacy, Chungbuk National University , Cheongju , Korea
| | - Kyungjae Kim
- a College of Pharmacy, Sahmyook University , Seoul , Korea
| |
Collapse
|
10
|
Lythgoe KA, Blanquart F, Pellis L, Fraser C. Large Variations in HIV-1 Viral Load Explained by Shifting-Mosaic Metapopulation Dynamics. PLoS Biol 2016; 14:e1002567. [PMID: 27706164 PMCID: PMC5051940 DOI: 10.1371/journal.pbio.1002567] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 09/08/2016] [Indexed: 12/17/2022] Open
Abstract
The viral population of HIV-1, like many pathogens that cause systemic infection, is structured and differentiated within the body. The dynamics of cellular immune trafficking through the blood and within compartments of the body has also received wide attention. Despite these advances, mathematical models, which are widely used to interpret and predict viral and immune dynamics in infection, typically treat the infected host as a well-mixed homogeneous environment. Here, we present mathematical, analytical, and computational results that demonstrate that consideration of the spatial structure of the viral population within the host radically alters predictions of previous models. We study the dynamics of virus replication and cytotoxic T lymphocytes (CTLs) within a metapopulation of spatially segregated patches, representing T cell areas connected by circulating blood and lymph. The dynamics of the system depend critically on the interaction between CTLs and infected cells at the within-patch level. We show that for a wide range of parameters, the system admits an unexpected outcome called the shifting-mosaic steady state. In this state, the whole body’s viral population is stable over time, but the equilibrium results from an underlying, highly dynamic process of local infection and clearance within T-cell centers. Notably, and in contrast to previous models, this new model can explain the large differences in set-point viral load (SPVL) observed between patients and their distribution, as well as the relatively low proportion of cells infected at any one time, and alters the predicted determinants of viral load variation. A novel metapopulation model of HIV suggests that within-host infections are characterized by a highly dynamic process of localized infection followed by clearance within T cell centers. When a person is infected with HIV, the initial peak level of virus in the blood is usually very high before a lower, relatively stable level is reached and maintained for the duration of the chronic infection. This stable level is known as the set-point viral load (SPVL) and is associated with severity of infection. SPVL is also highly variable among patients, ranging from 100 to a million copies of the virus per mL of blood. The replicative capacity of the infecting virus and the strength of the immune response both influence SPVL. However, standard mathematical models show that variation in these two factors cannot easily reproduce the observed distribution of SPVL among patients. Standard models typically treat infected individuals as well-mixed systems, but in reality viral replication is localised in T-cell centres, or patches, found in secondary lymphoid tissue. To account for this population structure, we developed a carefully parameterised metapopulation model. We find the system can reach a steady state at which the viral load in the blood is relatively stable, representing SPVL, but surprisingly, the patches are highly dynamic, characterised by bursts of infection followed by elimination of virus due to localised host immune responses. Significantly, this model can reproduce the wide distribution of SPVLs found among infected individuals for realistic distributions of viral replicative capacity and strength of immune response. Our model can also be used in the future to understand other aspects of chronic HIV infection.
Collapse
Affiliation(s)
- Katrina A. Lythgoe
- Department of Zoology, Tinbergen Building, University of Oxford, Oxford, United Kingdom
- Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, St. Mary’s Campus, London, United Kingdom
- * E-mail:
| | - François Blanquart
- Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, St. Mary’s Campus, London, United Kingdom
| | - Lorenzo Pellis
- Mathematics Institute, Zeeman Building, University of Warwick, Coventry, United Kingdom
| | - Christophe Fraser
- Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, St. Mary’s Campus, London, United Kingdom
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
11
|
Halle S, Keyser KA, Stahl FR, Busche A, Marquardt A, Zheng X, Galla M, Heissmeyer V, Heller K, Boelter J, Wagner K, Bischoff Y, Martens R, Braun A, Werth K, Uvarovskii A, Kempf H, Meyer-Hermann M, Arens R, Kremer M, Sutter G, Messerle M, Förster R. In Vivo Killing Capacity of Cytotoxic T Cells Is Limited and Involves Dynamic Interactions and T Cell Cooperativity. Immunity 2016; 44:233-45. [PMID: 26872694 PMCID: PMC4846978 DOI: 10.1016/j.immuni.2016.01.010] [Citation(s) in RCA: 172] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 10/08/2015] [Accepted: 11/18/2015] [Indexed: 01/11/2023]
Abstract
According to in vitro assays, T cells are thought to kill rapidly and efficiently, but the efficacy and dynamics of cytotoxic T lymphocyte (CTL)-mediated killing of virus-infected cells in vivo remains elusive. We used two-photon microscopy to quantify CTL-mediated killing in mice infected with herpesviruses or poxviruses. On average, one CTL killed 2–16 virus-infected cells per day as determined by real-time imaging and by mathematical modeling. In contrast, upon virus-induced MHC class I downmodulation, CTLs failed to destroy their targets. During killing, CTLs remained migratory and formed motile kinapses rather than static synapses with targets. Viruses encoding the calcium sensor GCaMP6s revealed strong heterogeneity in individual CTL functional capacity. Furthermore, the probability of death of infected cells increased for those contacted by more than two CTLs, indicative of CTL cooperation. Thus, direct visualization of CTLs during killing of virus-infected cells reveals crucial parameters of CD8+ T cell immunity. Two-photon imaging indicates that CTLs kill 2–16 virus-infected cells per day CTLs form kinapses rather than stable synapses when killing virus-infected cells Some CTL contacts trigger long-lasting calcium fluxes in virus-infected cells CTLs can cooperate during killing of virus-infected cells
Collapse
Affiliation(s)
- Stephan Halle
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany.
| | | | - Felix Rolf Stahl
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Andreas Busche
- Institute of Virology, Hannover Medical School, 30625 Hannover, Germany
| | - Anja Marquardt
- Institute of Virology, Hannover Medical School, 30625 Hannover, Germany
| | - Xiang Zheng
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Melanie Galla
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Vigo Heissmeyer
- Institute for Immunology, Ludwig-Maximilians-Universität München, 80336 München, Germany; Institute of Molecular Immunology, Helmholtz Zentrum München, 81377 München, Germany
| | - Katrin Heller
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Jasmin Boelter
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Karen Wagner
- Institute of Virology, Hannover Medical School, 30625 Hannover, Germany
| | - Yvonne Bischoff
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Rieke Martens
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Asolina Braun
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Kathrin Werth
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Alexey Uvarovskii
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Harald Kempf
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; Institute for Biochemistry, Biotechnology, and Bioinformatics, Technische Universität Braunschweig, 38124 Braunschweig, Germany
| | - Ramon Arens
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Melanie Kremer
- Institute for Infectious Diseases and Zoonoses, Ludwig-Maximilians-Universität München, 80539 München, Germany
| | - Gerd Sutter
- Institute for Infectious Diseases and Zoonoses, Ludwig-Maximilians-Universität München, 80539 München, Germany
| | - Martin Messerle
- Institute of Virology, Hannover Medical School, 30625 Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany.
| |
Collapse
|
12
|
Immune-surveillance through exhausted effector T-cells. Curr Opin Virol 2016; 16:49-54. [PMID: 26826950 DOI: 10.1016/j.coviro.2016.01.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 12/16/2015] [Accepted: 01/05/2016] [Indexed: 12/21/2022]
Abstract
Pathogens such as the human immunodeficiency virus (HIV), the hepatitis B and C virus (HBV, HCV) and certain strains of the rodent lymphocytic choriomeningitis virus (LCMV) establish a state of persisting viral replication. This occurs besides strong adoptive immune responses and the induction of large numbers of activated pathogen-specific T-cells. The failure of the immune system to clear these viruses is typically attributed to a loss of effector T-cell function-a phenomenon referred to as T-cell exhaustion. Though largely accepted, this loss of function concept is being more and more challenged by comprehensive clinical and experimental observations which highlight that T-cells in chronic infections are more functional than previously considered. Here, we highlight examples that demonstrate that such T-cells mediate a profound form of immune-surveillance. We also briefly discuss the opportunities and limitations of employing 'exhausted' T-cells for therapeutic purposes.
Collapse
|
13
|
Understanding Experimental LCMV Infection of Mice: The Role of Mathematical Models. J Immunol Res 2015; 2015:739706. [PMID: 26576439 PMCID: PMC4631900 DOI: 10.1155/2015/739706] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 09/27/2015] [Indexed: 12/28/2022] Open
Abstract
Virus infections represent complex biological systems governed by multiple-level regulatory processes of virus replication and host immune responses. Understanding of the infection means an ability to predict the systems behaviour under various conditions. Such predictions can only rely upon quantitative mathematical models. The model formulations should be tightly linked to a fundamental step called “coordinatization” (Hermann Weyl), that is, the definition of observables, parameters, and structures that enable the link with a biological phenotype. In this review, we analyse the mathematical modelling approaches to LCMV infection in mice that resulted in quantification of some fundamental parameters of the CTL-mediated virus control including the rates of T cell turnover, infected target cell elimination, and precursor frequencies. We show how the modelling approaches can be implemented to address diverse aspects of immune system functioning under normal conditions and in response to LCMV and, importantly, make quantitative predictions of the outcomes of immune system perturbations. This may highlight the notion that data-driven applications of meaningful mathematical models in infection biology remain a challenge.
Collapse
|