129 results on '"Zehn, D"'
Search Results
2. Perforin inhibition protects from lethal endothelial damage during fulminant viral hepatitis
- Author
-
Welz, M., Eickhoff, S., Abdullah, Z., Trebicka, J., Gartlan, K. H., Spicer, J. A., Demetris, A. J., Akhlaghi, H., Anton, M., Manske, K., Zehn, D., Nieswandt, B., Kurts, C., Trapani, J. A., Knolle, P., Wohlleber, D., and Kastenmüller, W.
- Published
- 2018
- Full Text
- View/download PDF
3. MYB orchestrates T cell exhaustion and response to checkpoint inhibition
- Author
-
Tsui, C, Kretschmer, L, Rapelius, S, Gabriel, SS, Chisanga, D, Knoepper, K, Utzschneider, DT, Nuessing, S, Liao, Y, Mason, T, Torres, SV, Wilcox, SA, Kanev, K, Jarosch, S, Leube, J, Nutt, SL, Zehn, D, Parish, IA, Kastenmueller, W, Shi, W, Buchholz, VR, Kallies, A, Tsui, C, Kretschmer, L, Rapelius, S, Gabriel, SS, Chisanga, D, Knoepper, K, Utzschneider, DT, Nuessing, S, Liao, Y, Mason, T, Torres, SV, Wilcox, SA, Kanev, K, Jarosch, S, Leube, J, Nutt, SL, Zehn, D, Parish, IA, Kastenmueller, W, Shi, W, Buchholz, VR, and Kallies, A
- Abstract
CD8+ T cells that respond to chronic viral infections or cancer are characterized by the expression of inhibitory receptors such as programmed cell death protein 1 (PD-1) and by the impaired production of cytokines. This state of restrained functionality-which is referred to as T cell exhaustion1,2-is maintained by precursors of exhausted T (TPEX) cells that express the transcription factor T cell factor 1 (TCF1), self-renew and give rise to TCF1- exhausted effector T cells3-6. Here we show that the long-term proliferative potential, multipotency and repopulation capacity of exhausted T cells during chronic infection are selectively preserved in a small population of transcriptionally distinct CD62L+ TPEX cells. The transcription factor MYB is not only essential for the development of CD62L+ TPEX cells and maintenance of the antiviral CD8+ T cell response, but also induces functional exhaustion and thereby prevents lethal immunopathology. Furthermore, the proliferative burst in response to PD-1 checkpoint inhibition originates exclusively from CD62L+ TPEX cells and depends on MYB. Our findings identify CD62L+ TPEX cells as a stem-like population that is central to the maintenance of long-term antiviral immunity and responsiveness to immunotherapy. Moreover, they show that MYB is a transcriptional orchestrator of two fundamental aspects of exhausted T cell responses: the downregulation of effector function and the long-term preservation of self-renewal capacity.
- Published
- 2022
4. Comparative single-cell trajectory network enrichment identifies pseudo-temporal systems biology patterns in hematopoiesis and CD8 T-cell development
- Author
-
Kristiyan Kanev, Jan Baumbach, Zehn D, Grønning Agb, Jesper Lund, Richard Röttger, Mhaned Oubounyt, and Tim Kacprowski
- Subjects
0303 health sciences ,Computer science ,Systems biology ,Single cell transcriptomics ,Cell ,Computational biology ,Expression (mathematics) ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Development (topology) ,Interaction network ,030220 oncology & carcinogenesis ,medicine ,Trajectory ,Cluster analysis ,030304 developmental biology - Abstract
Single cell transcriptomics (scRNA-seq) technologies allow for investigating cellular processes on an unprecedented resolution. While software packages for scRNA-seq raw data analysis exist, no method for the extraction of systems biology signatures that drive different pseudo-time trajectories exists. Hence, pseudo-temporal molecular sub-network expression profiles remain undetermined, thus, hampering our understanding of the molecular control of cellular development on a single cell resolution. We have developed Scellnetor, the first network-constraint time-series clustering algorithm implemented as interactive webtool to identify modules of genes connected in a molecular interaction network that show differentiating temporal expression patterns. Scellnetor allows selecting two differentiation courses or two developmental trajectories for comparison on a systems biology level. Scellnetor identifies mechanisms driving hematopoiesis in mouse and mechanistically interpretable subnetworks driving dysfunctional CD8 T-cell development in chronic infections. Scellnetor is the first method to allow for single cell trajectory network enrichment for systems level hypotheses generation, thus lifting scRNA-seq data analysis to a systems biology level. It is available as an interactive online tool at https://exbio.wzw.tum.de/scellnetor/.
- Published
- 2020
- Full Text
- View/download PDF
5. SARS-CoV-2 infections in cancer outpatients - Most infected patients are asymptomatic carriers without impact on chemotherapy
- Author
-
Hempel, L., Piehler, A., Pfaffl, M.W., Molnar, J., Kirchner, B., Robert, Sebastian, Veloso, J., Gandorfer, B., Trepotec, Z., Mederle, S., Keim, S., Milani, V., Ebner, F., Schweneker, K., Fleischmann, B., Kleespies, A., Scheiber, J., Hempel, D., Zehn, D., and Publica
- Abstract
Oncologic patients are regarded as the population most at risk of developing a severe course of COVID-19 due to the fact that malignant diseases and chemotherapy often weaken the immune system. In the face of the ongoing SARS-CoV-2 pandemic, how particular patients deal with this infection remains an important question. In the period between the 15 and 26 April 2020, a total of 1227 patients were tested in one of seven oncologic outpatient clinics for SARS-CoV-2, regardless of symptoms, employing RT-qPCR. Of 1227 patients, 78 (6.4%) were tested positive of SARS-CoV-2. Only one of the patients who tested positive developed a severe form of COVID-19 with pneumonia (CURB-65 score of 2), and two patients showed mild symptoms. Fourteen of 75 asymptomatic but positively tested patients received chemotherapy or chemo-immunotherapy according to their regular therapy algorithm (±4 weeks of SARS-CoV-2 test), and 48 of 78 (61.5%) positive-tested patients received glucocorticoids as co-medication. None of the asymptomatic infected patients showed unexpected complications due to the SARS-CoV-2 infection during the cancer treatment. These data clearly contrast the view that patients with an oncologic disease are particularly vulnerable to SARS-CoV-2 and suggest that compromising therapies could be continued or started despite the ongoing pandemic. Moreover the relatively low appearance of symptoms due to COVID-19 among patients on chemotherapy and other immunosuppressive co-medication like glucocorticoids indicate that suppressing the response capacity of the immune system reduces disease severity.
- Published
- 2020
6. PTPN2 Deficiency Enhances Programmed T Cell Expansion and Survival Capacity of Activated T Cells
- Author
-
Flosbach, M, Oberle, SG, Scherer, S, Zecha, J, von Hoesslin, M, Wiede, F, Chennupati, V, Cullen, JG, List, M, Pauling, JK, Baumbach, J, Kuster, B, Tiganis, T, Zehn, D, Flosbach, M, Oberle, SG, Scherer, S, Zecha, J, von Hoesslin, M, Wiede, F, Chennupati, V, Cullen, JG, List, M, Pauling, JK, Baumbach, J, Kuster, B, Tiganis, T, and Zehn, D
- Abstract
Manipulating molecules that impact T cell receptor (TCR) or cytokine signaling, such as the protein tyrosine phosphatase non-receptor type 2 (PTPN2), has significant potential for advancing T cell-based immunotherapies. Nonetheless, it remains unclear how PTPN2 impacts the activation, survival, and memory formation of T cells. We find that PTPN2 deficiency renders cells in vivo and in vitro less dependent on survival-promoting cytokines, such as interleukin (IL)-2 and IL-15. Remarkably, briefly ex vivo-activated PTPN2-deficient T cells accumulate in 3- to 11-fold higher numbers following transfer into unmanipulated, antigen-free mice. Moreover, the absence of PTPN2 augments the survival of short-lived effector T cells and allows them to robustly re-expand upon secondary challenge. Importantly, we find no evidence for impaired effector function or memory formation. Mechanistically, PTPN2 deficiency causes broad changes in the expression and phosphorylation of T cell expansion and survival-associated proteins. Altogether, our data underline the therapeutic potential of targeting PTPN2 in T cell-based therapies to augment the number and survival capacity of antigen-specific T cells.
- Published
- 2020
7. 1680P SARS-CoV-2 infections in outpatients with cancer: Most infected patients are asymptomatic carriers without impact on chemotherapy
- Author
-
Hempel, D., primary, Milani, V., additional, Kleespies, A., additional, Hempel, L., additional, Ebner, F., additional, Zehn, D., additional, and Keim, S., additional
- Published
- 2020
- Full Text
- View/download PDF
8. MiR-23∼27∼24-mediated control of humoral immunity reveals a TOX-driven regulatory circuit in follicular helper T cell differentiation
- Author
-
Wu, C-J, Cho, S, Huang, H-Y, Lu, C-H, Russ, J, Cruz, LO, da Cunha, FF, Chen, M-C, Lin, L-L, Warner, LM, Liao, H-K, Utzschneider, DT, Quon, S, Berner, J, Saraiva Camara, NO, Zehn, D, Belmonte, JCI, Chen, L-C, Huang, S-F, Kuo, M-L, Lu, L-F, Wu, C-J, Cho, S, Huang, H-Y, Lu, C-H, Russ, J, Cruz, LO, da Cunha, FF, Chen, M-C, Lin, L-L, Warner, LM, Liao, H-K, Utzschneider, DT, Quon, S, Berner, J, Saraiva Camara, NO, Zehn, D, Belmonte, JCI, Chen, L-C, Huang, S-F, Kuo, M-L, and Lu, L-F
- Abstract
Follicular helper T (TFH) cells are essential for generating protective humoral immunity. To date, microRNAs (miRNAs) have emerged as important players in regulating TFH cell biology. Here, we show that loss of miR-23~27~24 clusters in T cells resulted in elevated TFH cell frequencies upon different immune challenges, whereas overexpression of this miRNA family led to reduced TFH cell responses. Mechanistically, miR-23~27~24 clusters coordinately control TFH cells through targeting a network of genes that are crucial for TFH cell biology. Among them, thymocyte selection-associated HMG-box protein (TOX) was identified as a central transcription regulator in TFH cell development. TOX is highly up-regulated in both mouse and human TFH cells in a BCL6-dependent manner. In turn, TOX promotes the expression of multiple molecules that play critical roles in TFH cell differentiation and function. Collectively, our results establish a key miRNA regulon that maintains optimal TFH cell responses for resultant humoral immunity.
- Published
- 2019
9. Perforin inhibition protects from lethal endothelial damage during fulminant viral hepatitis
- Author
-
Welz, M, Eickhoff, S, Abdullah, Z, Trebicka, J, Gartlan, KH, Spicer, JA, Demetris, AJ, Akhlaghi, H, Anton, M, Manske, K, Zehn, D, Nieswandt, B, Kurts, C, Trapani, JA, Knolle, P, Wohlleber, D, Kastenmueller, W, Welz, M, Eickhoff, S, Abdullah, Z, Trebicka, J, Gartlan, KH, Spicer, JA, Demetris, AJ, Akhlaghi, H, Anton, M, Manske, K, Zehn, D, Nieswandt, B, Kurts, C, Trapani, JA, Knolle, P, Wohlleber, D, and Kastenmueller, W
- Abstract
CD8 T cells protect the liver against viral infection, but can also cause severe liver damage that may even lead to organ failure. Given the lack of mechanistic insights and specific treatment options in patients with acute fulminant hepatitis, we develop a mouse model reflecting a severe acute virus-induced CD8 T cell-mediated hepatitis. Here we show that antigen-specific CD8 T cells induce liver damage in a perforin-dependent manner, yet liver failure is not caused by effector responses targeting virus-infected hepatocytes alone. Additionally, CD8 T cell mediated elimination of cross-presenting liver sinusoidal endothelial cells causes endothelial damage that leads to a dramatically impaired sinusoidal perfusion and indirectly to hepatocyte death. With the identification of perforin-mediated killing as a critical pathophysiologic mechanism of liver failure and the protective function of a new class of perforin inhibitor, our study opens new potential therapeutic angles for fulminant viral hepatitis.
- Published
- 2018
10. NLRC5 shields T lymphocytes from NK-cell-mediated elimination under inflammatory conditions
- Author
-
Ludigs, K, Jandus, C, Utzschneider, DT, Staehli, F, Bessoles, S, Dang, AT, Rota, G, Castro, W, Zehn, D, Vivier, E, Held, W, Romero, P, Guarda, G, Ludigs, K, Jandus, C, Utzschneider, DT, Staehli, F, Bessoles, S, Dang, AT, Rota, G, Castro, W, Zehn, D, Vivier, E, Held, W, Romero, P, and Guarda, G
- Abstract
NLRC5 is a transcriptional regulator of MHC class I (MHCI), which maintains high MHCI expression particularly in T cells. Recent evidence highlights an important NK-T-cell crosstalk, raising the question on whether NLRC5 specifically modulates this interaction. Here we show that NK cells from Nlrc5-deficient mice exhibit moderate alterations in inhibitory receptor expression and responsiveness. Interestingly, NLRC5 expression in T cells is required to protect them from NK-cell-mediated elimination upon inflammation. Using T-cell-specific Nlrc5-deficient mice, we show that NK cells surprisingly break tolerance even towards 'self' Nlrc5-deficient T cells under inflammatory conditions. Furthermore, during chronic LCMV infection, the total CD8(+) T-cell population is severely decreased in these mice, a phenotype reverted by NK-cell depletion. These findings strongly suggest that endogenous T cells with low MHCI expression become NK-cell targets, having thus important implications for T-cell responses in naturally or therapeutically induced inflammatory conditions.
- Published
- 2016
11. High antigen levels induce an exhausted phenotype in a chronic infection without impairing T cell expansion and survival
- Author
-
Utzschneider, DT, Alfei, F, Roelli, P, Barras, D, Chennupati, V, Darbre, S, Delorenzi, M, Pinschewer, DD, Zehn, D, Utzschneider, DT, Alfei, F, Roelli, P, Barras, D, Chennupati, V, Darbre, S, Delorenzi, M, Pinschewer, DD, and Zehn, D
- Abstract
Chronic infections induce T cells showing impaired cytokine secretion and up-regulated expression of inhibitory receptors such as PD-1. What determines the acquisition of this chronic phenotype and how it impacts T cell function remain vaguely understood. Using newly generated recombinant antigen variant-expressing chronic lymphocytic choriomeningitis virus (LCMV) strains, we uncovered that T cell differentiation and acquisition of a chronic or exhausted phenotype depend critically on the frequency of T cell receptor (TCR) engagement and less significantly on the strength of TCR stimulation. In fact, we noted that low-level antigen exposure promotes the formation of T cells with an acute phenotype in chronic infections. Unexpectedly, we found that T cell populations with an acute or chronic phenotype are maintained equally well in chronic infections and undergo comparable primary and secondary expansion. Thus, our observations contrast with the view that T cells with a typical chronic infection phenotype are severely functionally impaired and rapidly transition into a terminal stage of differentiation. Instead, our data unravel that T cells primarily undergo a form of phenotypic and functional differentiation in the early phase of a chronic LCMV infection without inheriting a net survival or expansion deficit, and we demonstrate that the acquired chronic phenotype transitions into the memory T cell compartment.
- Published
- 2016
12. A TCF1 expressing memory-like population of Hepatitis C virus-specific CD8+ T cells is maintained after DAA-mediated viral elimination
- Author
-
Wieland, D, additional, Kemming, J, additional, Schuch, A, additional, Neumann-Haefelin, C, additional, Knolle, P, additional, Held, W, additional, Zehn, D, additional, Hofmann, M, additional, and Thimme, R, additional
- Published
- 2016
- Full Text
- View/download PDF
13. A Memory-Like Subset Dominates the HCV-Specific CD8+ T-Cell Compartment after DAA-Mediated Antigen Removal
- Author
-
Wieland, D., primary, Schuch, A., additional, Held, W., additional, Zehn, D., additional, Hofmann, M., additional, and Thimme, R., additional
- Published
- 2016
- Full Text
- View/download PDF
14. Molecular insights for optimizing T cell receptor specificity against cancer
- Author
-
Hebeisen, M., Oberle, S.G., Presotto, D., Speiser, D.E., Zehn, D., and Rufer, N.
- Subjects
chemical and pharmacologic phenomena - Abstract
Cytotoxic CD8 T cells mediate immunity to pathogens and they are able to eliminate malignant cells. Immunity to viruses and bacteria primarily involves CD8 T cells bearing high affinity T cell receptors (TCRs), which are specific to pathogen-derived (non-self) antigens. Given the thorough elimination of high affinity self/tumor-antigen reactive T cells by central and peripheral tolerance mechanisms, anti-cancer immunity mostly depends on TCRs with intermediate-to-low affinity for self-antigens. Because of this, a promising novel therapeutic approach to increase the efficacy of tumor-reactive T cells is to engineer their TCRs, with the aim to enhance their binding kinetics to pMHC complexes, or to directly manipulate the TCR-signaling cascades. Such manipulations require a detailed knowledge on how pMHC-TCR and co-receptors binding kinetics impact the T cell response. In this review, we present the current knowledge in this field. We discuss future challenges in identifying and targeting the molecular mechanisms to enhance the function of natural or TCR-affinity optimized T cells, and we provide perspectives for the development of protective anti-tumor T cell responses.
- Published
- 2013
15. Functional Avidity: A Measure to Predict the Efficacy of Effector T Cells?
- Author
-
Viganò, S., Utzschneider, D.T., Perreau, M., Pantaleo, G., Zehn, D., and Harari, A.
- Subjects
Article Subject ,chemical and pharmacologic phenomena - Abstract
The functional avidity is determined by exposing T-cell populations in vitro to different amounts of cognate antigen. T-cells with high functional avidity respond to low antigen doses. This in vitro measure is thought to correlate well with the in vivo effector capacity of T-cells. We here present the multifaceted factors determining and influencing the functional avidity of T-cells. We outline how changes in the functional avidity can occur over the course of an infection. This process, known as avidity maturation, can occur despite the fact that T-cells express a fixed TCR. Furthermore, examples are provided illustrating the importance of generating T-cell populations that exhibit a high functional avidity when responding to an infection or tumors. Furthermore, we discuss whether criteria based on which we evaluate an effective T-cell response to acute infections can also be applied to chronic infections such as HIV. Finally, we also focus on observations that high-avidity T-cells show higher signs of exhaustion and facilitate the emergence of virus escape variants. The review summarizes our current understanding of how this may occur as well as how T-cells of different functional avidity contribute to antiviral and anti-tumor immunity. Enhancing our knowledge in this field is relevant for tumor immunotherapy and vaccines design.
- Published
- 2012
- Full Text
- View/download PDF
16. TCR signaling requirements for activating T cells and for generating memory
- Author
-
Zehn, D., King, C., Bevan, M.J., and Palmer, E.
- Subjects
Immunologic Memory ,Lymphocyte Activation ,Major Histocompatibility Complex/physiology ,Receptors, Antigen, T-Cell/metabolism ,Signal Transduction ,T-Lymphocytes/immunology - Abstract
Over the last two decades the molecular and cellular mechanisms underlying T cell activation, expansion, differentiation, and memory formation have been intensively investigated. These studies revealed that the generation of memory T cells is critically impacted by a number of factors, including the magnitude of the inflammatory response and cytokine production, the type of dendritic cell [DC] that presents the pathogen derived antigen, their maturation status, and the concomitant provision of costimulation. Nevertheless, the primary stimulus leading to T cell activation is generated through the T cell receptor [TCR] following its engagement with a peptide MHC ligand [pMHC]. The purpose of this review is to highlight classical and recent findings on how antigen recognition, the degree of TCR stimulation, and intracellular signal transduction pathways impact the formation of effector and memory T cells.
- Published
- 2012
17. Functional Avidity: A Measure to Predict the Efficacy of Effector T Cells?
- Author
-
Vigano, S, Utzschneider, DT, Perreau, M, Pantaleo, G, Zehn, D, Harari, A, Vigano, S, Utzschneider, DT, Perreau, M, Pantaleo, G, Zehn, D, and Harari, A
- Abstract
The functional avidity is determined by exposing T-cell populations in vitro to different amounts of cognate antigen. T-cells with high functional avidity respond to low antigen doses. This in vitro measure is thought to correlate well with the in vivo effector capacity of T-cells. We here present the multifaceted factors determining and influencing the functional avidity of T-cells. We outline how changes in the functional avidity can occur over the course of an infection. This process, known as avidity maturation, can occur despite the fact that T-cells express a fixed TCR. Furthermore, examples are provided illustrating the importance of generating T-cell populations that exhibit a high functional avidity when responding to an infection or tumors. Furthermore, we discuss whether criteria based on which we evaluate an effective T-cell response to acute infections can also be applied to chronic infections such as HIV. Finally, we also focus on observations that high-avidity T-cells show higher signs of exhaustion and facilitate the emergence of virus escape variants. The review summarizes our current understanding of how this may occur as well as how T-cells of different functional avidity contribute to antiviral and anti-tumor immunity. Enhancing our knowledge in this field is relevant for tumor immunotherapy and vaccines design.
- Published
- 2012
18. SAT-133 - A Memory-Like Subset Dominates the HCV-Specific CD8+ T-Cell Compartment after DAA-Mediated Antigen Removal
- Author
-
Wieland, D., Schuch, A., Held, W., Zehn, D., Hofmann, M., and Thimme, R.
- Published
- 2016
- Full Text
- View/download PDF
19. One naive T cell, multiple fates in CD8+ T cell differentiation
- Author
-
Gerlach, C., van Heijst, J.W.J., Swart, E., Sie, D., Armstrong, N., Kerkhoven, R.M., Zehn, D., Bevan, M.J., Schepers, K., Schumacher, T.N.M., Gerlach, C., van Heijst, J.W.J., Swart, E., Sie, D., Armstrong, N., Kerkhoven, R.M., Zehn, D., Bevan, M.J., Schepers, K., and Schumacher, T.N.M.
- Abstract
The mechanism by which the immune system produces effector and memory T cells is largely unclear. To allow a large-scale assessment of the development of single naive T cells into different subsets, we have developed a technology that introduces unique genetic tags (barcodes) into naive T cells. By comparing the barcodes present in antigen-specific effector and memory T cell populations in systemic and local infection models, at different anatomical sites, and for TCR–pMHC interactions of different avidities, we demonstrate that under all conditions tested, individual naive T cells yield both effector and memory CD8+ T cell progeny. This indicates that effector and memory fate decisions are not determined by the nature of the priming antigen-presenting cell or the time of T cell priming. Instead, for both low and high avidity T cells, individual naive T cells have multiple fates and can differentiate into effector and memory T cell subsets.
- Published
- 2010
20. Functional avidity: a measure to predict the efficacy of effector T cells?
- Author
-
Viganò S, Utzschneider DT, Perreau M, Pantaleo G, Zehn D, and Harari A
- Subjects
chemical and pharmacologic phenomena - Abstract
The functional avidity is determined by exposing T cell populations in vitro to different amounts of cognate antigen. T cells with high functional avidity respond to low antigen doses. This in vitro measure is thought to correlate well with the in vivo effector capacity of T cells. We here present the multifaceted factors determining and influencing the functional avidity of T cells. We outline how changes in the functional avidity can occur over the course of an infection. This process known as avidity maturation can occur despite the fact that T cells express a fixed TCR. Furthermore examples are provided illustrating the importance of generating T cell populations that exhibit a high functional avidity when responding to an infection or tumors. Furthermore we discuss whether criteria based on which we evaluate an effective T cell response to acute infections can also be applied to chronic infections such as HIV. Finally we also focus on observations that high avidity T cells show higher signs of exhaustion and facilitate the emergence of virus escape variants. The review summarizes our current understanding of how this may occur as well as how T cells of different functional avidity contribute to antiviral
- Published
- 2013
21. Efficiency of peptide presentation by dendritic cells compared with other cell types: implications for cross-priming
- Author
-
Zehn, D., primary, Cohen, C. J., additional, Reiter, Y., additional, and Walden, P., additional
- Published
- 2006
- Full Text
- View/download PDF
22. Dynamic landscapes and protective immunity coordinated by influenza-specific lung-resident memory CD8 + T cells revealed by intravital imaging.
- Author
-
van de Wall S, Anthony SM, Hancox LS, Pewe LL, Langlois RA, Zehn D, Badovinac VP, and Harty JT
- Subjects
- Animals, Mice, Mice, Inbred C57BL, Interferon-gamma metabolism, Interferon-gamma immunology, Intravital Microscopy, Monocytes immunology, Lung immunology, Lung virology, Orthomyxoviridae Infections immunology, CD8-Positive T-Lymphocytes immunology, Immunologic Memory immunology, Integrin alpha Chains metabolism, Influenza A virus immunology, Antigens, CD metabolism, Memory T Cells immunology
- Abstract
Lung-tissue-resident memory (T
RM ) CD8+ T cells are critical for heterosubtypic immunity against influenza virus (IAV) reinfection. How TRM cells surveil the lung, respond to infection, and interact with other cells remains unresolved. Here, we used IAV infection of mice in combination with intravital and static imaging to define the spatiotemporal dynamics of lung TRM cells before and after recall infection. CD69+ CD103+ TRM cells preferentially localized to lung sites of prior IAV infection, where they exhibited patrolling behavior. After rechallenge, lung TRM cells formed tight clusters in an antigen-dependent manner. Transcriptomic analysis of IAV-specific TRM cells revealed the expression of several factors that regulate myeloid cell biology. In vivo rechallenge experiments demonstrated that protection elicited by TRM cells is orchestrated in part by interferon (IFN)-γ-mediated recruitment of inflammatory monocytes into the lungs. Overall, these data illustrate the dynamic landscapes of CD103+ lung TRM cells that mediate early protective immunity against IAV infection., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
23. T cell dysfunction and therapeutic intervention in cancer.
- Author
-
Zebley CC, Zehn D, Gottschalk S, and Chi H
- Subjects
- Humans, Animals, CD8-Positive T-Lymphocytes immunology, Tumor Escape, Cell Differentiation immunology, Neoplasms immunology, Neoplasms therapy, Tumor Microenvironment immunology, Immunotherapy methods
- Abstract
Recent advances in immunotherapy have affirmed the curative potential of T cell-based approaches for treating relapsed and refractory cancers. However, the therapeutic efficacy is limited in part owing to the ability of cancers to evade immunosurveillance and adapt to immunological pressure. In this Review, we provide a brief overview of cancer-mediated immunosuppressive mechanisms with a specific focus on the repression of the surveillance and effector function of T cells. We discuss CD8
+ T cell exhaustion and functional heterogeneity and describe strategies for targeting the molecular checkpoints that restrict T cell differentiation and effector function to bolster immunotherapeutic effects. We also delineate the emerging contributions of the tumor microenvironment to T cell metabolism and conclude by highlighting discovery-based approaches for developing future cellular therapies. Continued exploration of T cell biology and engineering hold great promise for advancing therapeutic interventions for cancer., (© 2024. Springer Nature America, Inc.)- Published
- 2024
- Full Text
- View/download PDF
24. A liver immune rheostat regulates CD8 T cell immunity in chronic HBV infection.
- Author
-
Bosch M, Kallin N, Donakonda S, Zhang JD, Wintersteller H, Hegenbarth S, Heim K, Ramirez C, Fürst A, Lattouf EI, Feuerherd M, Chattopadhyay S, Kumpesa N, Griesser V, Hoflack JC, Siebourg-Polster J, Mogler C, Swadling L, Pallett LJ, Meiser P, Manske K, de Almeida GP, Kosinska AD, Sandu I, Schneider A, Steinbacher V, Teng Y, Schnabel J, Theis F, Gehring AJ, Boonstra A, Janssen HLA, Vandenbosch M, Cuypers E, Öllinger R, Engleitner T, Rad R, Steiger K, Oxenius A, Lo WL, Klepsch V, Baier G, Holzmann B, Maini MK, Heeren R, Murray PJ, Thimme R, Herrmann C, Protzer U, Böttcher JP, Zehn D, Wohlleber D, Lauer GM, Hofmann M, Luangsay S, and Knolle PA
- Subjects
- Animals, Humans, Male, Mice, Cyclic AMP Response Element Modulator metabolism, Cyclic AMP-Dependent Protein Kinases metabolism, Hepatitis B virus immunology, Hepatocytes immunology, Hepatocytes virology, Phosphorylation, Signal Transduction, Lymphocyte Activation, CD8-Positive T-Lymphocytes enzymology, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes pathology, Hepatitis B, Chronic immunology, Hepatitis B, Chronic virology, Liver immunology, Liver virology
- Abstract
Chronic hepatitis B virus (HBV) infection affects 300 million patients worldwide
1,2 , in whom virus-specific CD8 T cells by still ill-defined mechanisms lose their function and cannot eliminate HBV-infected hepatocytes3-7 . Here we demonstrate that a liver immune rheostat renders virus-specific CD8 T cells refractory to activation and leads to their loss of effector functions. In preclinical models of persistent infection with hepatotropic viruses such as HBV, dysfunctional virus-specific CXCR6+ CD8 T cells accumulated in the liver and, as a characteristic hallmark, showed enhanced transcriptional activity of cAMP-responsive element modulator (CREM) distinct from T cell exhaustion. In patients with chronic hepatitis B, circulating and intrahepatic HBV-specific CXCR6+ CD8 T cells with enhanced CREM expression and transcriptional activity were detected at a frequency of 12-22% of HBV-specific CD8 T cells. Knocking out the inhibitory CREM/ICER isoform in T cells, however, failed to rescue T cell immunity. This indicates that CREM activity was a consequence, rather than the cause, of loss in T cell function, further supported by the observation of enhanced phosphorylation of protein kinase A (PKA) which is upstream of CREM. Indeed, we found that enhanced cAMP-PKA-signalling from increased T cell adenylyl cyclase activity augmented CREM activity and curbed T cell activation and effector function in persistent hepatic infection. Mechanistically, CD8 T cells recognizing their antigen on hepatocytes established close and extensive contact with liver sinusoidal endothelial cells, thereby enhancing adenylyl cyclase-cAMP-PKA signalling in T cells. In these hepatic CD8 T cells, which recognize their antigen on hepatocytes, phosphorylation of key signalling kinases of the T cell receptor signalling pathway was impaired, which rendered them refractory to activation. Thus, close contact with liver sinusoidal endothelial cells curbs the activation and effector function of HBV-specific CD8 T cells that target hepatocytes expressing viral antigens by means of the adenylyl cyclase-cAMP-PKA axis in an immune rheostat-like fashion., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
25. Antitumor progenitor exhausted CD8 + T cells are sustained by TCR engagement.
- Author
-
Lan X, Mi T, Alli S, Guy C, Djekidel MN, Liu X, Boi S, Chowdhury P, He M, Zehn D, Feng Y, and Youngblood B
- Subjects
- Animals, Mice, Cell Differentiation immunology, Dendritic Cells immunology, Signal Transduction immunology, Mice, Knockout, Lymphocyte Activation immunology, Cell Self Renewal, Mice, Transgenic, Early Growth Response Protein 2, CD8-Positive T-Lymphocytes immunology, Receptors, Antigen, T-Cell metabolism, Receptors, Antigen, T-Cell immunology, Carcinoma, Lewis Lung immunology, Carcinoma, Lewis Lung pathology, Carcinoma, Lewis Lung metabolism, Mice, Inbred C57BL, Hepatocyte Nuclear Factor 1-alpha metabolism
- Abstract
The durability of an antitumor immune response is mediated in part by the persistence of progenitor exhausted CD8
+ T cells (Tpex). Tpex serve as a resource for replenishing effector T cells and preserve their quantity through self-renewal. However, it is unknown how T cell receptor (TCR) engagement affects the self-renewal capacity of Tpex in settings of continued antigen exposure. Here we use a Lewis lung carcinoma model that elicits either optimal or attenuated TCR signaling in CD8+ T cells to show that formation of Tpex in tumor-draining lymph nodes and their intratumoral persistence is dependent on optimal TCR engagement. Notably, attenuated TCR stimulation accelerates the terminal differentiation of optimally primed Tpex. This TCR-reinforced Tpex development and self-renewal is coupled to proximal positioning to dendritic cells and epigenetic imprinting involving increased chromatin accessibility at Egr2 and Tcf1 target loci. Collectively, this study highlights the critical function of TCR engagement in sustaining Tpex during tumor progression., (© 2024. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2024
- Full Text
- View/download PDF
26. PGE 2 limits effector expansion of tumour-infiltrating stem-like CD8 + T cells.
- Author
-
Lacher SB, Dörr J, de Almeida GP, Hönninger J, Bayerl F, Hirschberger A, Pedde AM, Meiser P, Ramsauer L, Rudolph TJ, Spranger N, Morotti M, Grimm AJ, Jarosch S, Oner A, Gregor L, Lesch S, Michaelides S, Fertig L, Briukhovetska D, Majed L, Stock S, Busch DH, Buchholz VR, Knolle PA, Zehn D, Dangaj Laniti D, Kobold S, and Böttcher JP
- Subjects
- Animals, Female, Humans, Male, Mice, Cell Differentiation, Cell Line, Tumor, Disease Models, Animal, Hepatocyte Nuclear Factor 1-alpha metabolism, Interleukin-2, Lymph Nodes cytology, Lymph Nodes immunology, Mice, Inbred C57BL, Receptors, Prostaglandin E, EP2 Subtype deficiency, Receptors, Prostaglandin E, EP2 Subtype metabolism, Receptors, Prostaglandin E, EP4 Subtype deficiency, Receptors, Prostaglandin E, EP4 Subtype metabolism, Signal Transduction, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Cell Proliferation, Dinoprostone metabolism, Lymphocytes, Tumor-Infiltrating cytology, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Neoplasms immunology, Neoplasms prevention & control, Stem Cells cytology, Stem Cells immunology, Stem Cells metabolism, Tumor Escape immunology
- Abstract
Cancer-specific TCF1
+ stem-like CD8+ T cells can drive protective anticancer immunity through expansion and effector cell differentiation1-4 ; however, this response is dysfunctional in tumours. Current cancer immunotherapies2,5-9 can promote anticancer responses through TCF1+ stem-like CD8+ T cells in some but not all patients. This variation points towards currently ill-defined mechanisms that limit TCF1+ CD8+ T cell-mediated anticancer immunity. Here we demonstrate that tumour-derived prostaglandin E2 (PGE2 ) restricts the proliferative expansion and effector differentiation of TCF1+ CD8+ T cells within tumours, which promotes cancer immune escape. PGE2 does not affect the priming of TCF1+ CD8+ T cells in draining lymph nodes. PGE2 acts through EP2 and EP4 (EP2 /EP4 ) receptor signalling in CD8+ T cells to limit the intratumoural generation of early and late effector T cell populations that originate from TCF1+ tumour-infiltrating CD8+ T lymphocytes (TILs). Ablation of EP2 /EP4 signalling in cancer-specific CD8+ T cells rescues their expansion and effector differentiation within tumours and leads to tumour elimination in multiple mouse cancer models. Mechanistically, suppression of the interleukin-2 (IL-2) signalling pathway underlies the PGE2 -mediated inhibition of TCF1+ TIL responses. Altogether, we uncover a key mechanism that restricts the IL-2 responsiveness of TCF1+ TILs and prevents anticancer T cell responses that originate from these cells. This study identifies the PGE2 -EP2 /EP4 axis as a molecular target to restore IL-2 responsiveness in anticancer TILs to achieve cancer immune control., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
27. Stat5a sobers up inTOXicated T cells.
- Author
-
Donhauser LV and Zehn D
- Subjects
- Humans, STAT5 Transcription Factor metabolism, Cell Differentiation, T-Lymphocytes metabolism, Neoplasms
- Abstract
Exhausted T cells are largely hampered by epigenetically enforced mechanisms that limit their effector potential. In this issue of Immunity, Beltra et al. found that Stat5 can alter these epigenetic profiles when T cells transition from the Tpex precursor stage into differentiated cells. At this stage, enforced Stat5 expression increases the number of intermediate exhausted T cells and induces durable effector cells with superior anti-tumor activity., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
28. An adrenaline kick to exhaust T cells.
- Author
-
Berner J and Zehn D
- Subjects
- Epinephrine, T-Lymphocytes
- Published
- 2023
- Full Text
- View/download PDF
29. Responses of patients with cancer to mRNA vaccines depend on the time interval between vaccination and last treatment.
- Author
-
Donhauser LV, Veloso de Oliveira J, Schick C, Manlik W, Styblova S, Lutzenberger S, Aigner M, Philipp P, Robert S, Gandorfer B, Hempel D, Hempel L, and Zehn D
- Subjects
- Humans, Vaccination, Medical Oncology, COVID-19 prevention & control, Neoplasms drug therapy, Hematologic Neoplasms
- Abstract
Background: Personalized mRNA vaccines are promising new therapeutic options for patients with cancer. Because mRNA vaccines are not yet approved for first-line therapy, the vaccines are presently applied to individuals that received prior therapies that can have immunocompromising effects. There is a need to address how prior treatments impact mRNA vaccine outcomes., Method: Therefore, we analyzed the response to BioNTech/Pfizer's anti-SARS-CoV-2 mRNA vaccine in 237 oncology outpatients, which cover a broad spectrum of hematologic malignancies and solid tumors and a variety of treatments. Patients were stratified by the time interval between the last treatment and first vaccination and by the presence or absence of florid tumors and IgG titers and T cell responses were analyzed 14 days after the second vaccination., Results: Regardless of the last treatment time point, our data indicate that vaccination responses in patients with checkpoint inhibition were comparable to healthy controls. In contrast, patients after chemotherapy or cortisone therapy did not develop an immune response until 6 months after the last systemic therapy and patients after Cht-immune checkpoint inhibitor and tyrosine kinase inhibitor therapy only after 12 months., Conclusion: Accordingly, our data support that timing of mRNA-based therapy is critical and we suggest that at least a 6-months or 12-months waiting interval should be observed before mRNA vaccination in systemically treated patients., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
30. The alarmin interleukin-33 promotes the expansion and preserves the stemness of Tcf-1 + CD8 + T cells in chronic viral infection.
- Author
-
Marx AF, Kallert SM, Brunner TM, Villegas JA, Geier F, Fixemer J, Abreu-Mota T, Reuther P, Bonilla WV, Fadejeva J, Kreutzfeldt M, Wagner I, Aparicio-Domingo P, Scarpellino L, Charmoy M, Utzschneider DT, Hagedorn C, Lu M, Cornille K, Stauffer K, Kreppel F, Merkler D, Zehn D, Held W, Luther SA, Löhning M, and Pinschewer DD
- Subjects
- Animals, Mice, Alarmins metabolism, Interleukin-1 Receptor-Like 1 Protein metabolism, Lymphocytic choriomeningitis virus, Mice, Inbred C57BL, Persistent Infection, T Cell Transcription Factor 1 metabolism, CD8-Positive T-Lymphocytes, Interleukin-33 metabolism, Lymphocytic Choriomeningitis immunology
- Abstract
T cell factor 1 (Tcf-1) expressing CD8
+ T cells exhibit stem-like self-renewing capacity, rendering them key for immune defense against chronic viral infection and cancer. Yet, the signals that promote the formation and maintenance of these stem-like CD8+ T cells (CD8+ SL) remain poorly defined. Studying CD8+ T cell differentiation in mice with chronic viral infection, we identified the alarmin interleukin-33 (IL-33) as pivotal for the expansion and stem-like functioning of CD8+ SL as well as for virus control. IL-33 receptor (ST2)-deficient CD8+ T cells exhibited biased end differentiation and premature loss of Tcf-1. ST2-deficient CD8+ SL responses were restored by blockade of type I interferon signaling, suggesting that IL-33 balances IFN-I effects to control CD8+ SL formation in chronic infection. IL-33 signals broadly augmented chromatin accessibility in CD8+ SL and determined these cells' re-expansion potential. Our study identifies the IL-33-ST2 axis as an important CD8+ SL-promoting pathway in the context of chronic viral infection., Competing Interests: Declaration of interests D.D.P. is a founder, consultant, and shareholder of Hookipa Pharma Inc. commercializing arenavirus-based vector technology, and he as well as W.V.B., S.M.K., and D.M. are listed as inventor on corresponding patents., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
31. A single-amino acid substitution in the adaptor LAT accelerates TCR proofreading kinetics and alters T-cell selection, maintenance and function.
- Author
-
Lo WL, Kuhlmann M, Rizzuto G, Ekiz HA, Kolawole EM, Revelo MP, Andargachew R, Li Z, Tsai YL, Marson A, Evavold BD, Zehn D, and Weiss A
- Subjects
- Mice, Animals, Amino Acid Substitution, Receptors, Antigen, T-Cell metabolism, Lymphocyte Activation, Phosphorylation, Phosphoproteins genetics, Adaptor Proteins, Signal Transducing metabolism, T-Lymphocytes
- Abstract
Mature T cells must discriminate between brief interactions with self-peptides and prolonged binding to agonists. The kinetic proofreading model posits that certain T-cell antigen receptor signaling nodes serve as molecular timers to facilitate such discrimination. However, the physiological significance of this regulatory mechanism and the pathological consequences of disrupting it are unknown. Here we report that accelerating the normally slow phosphorylation of the linker for activation of T cells (LAT) residue Y136 by introducing an adjacent Gly135Asp alteration (LAT
G135D ) disrupts ligand discrimination in vivo. The enhanced self-reactivity of LATG135D T cells triggers excessive thymic negative selection and promotes T-cell anergy. During Listeria infection, LATG135D T cells expand more than wild-type counterparts in response to very weak stimuli but display an imbalance between effector and memory responses. Moreover, despite their enhanced engagement of central and peripheral tolerance mechanisms, mice bearing LATG135D show features associated with autoimmunity and immunopathology. Our data reveal the importance of kinetic proofreading in balancing tolerance and immunity., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
32. Pyrimidine de novo synthesis inhibition selectively blocks effector but not memory T cell development.
- Author
-
Scherer S, Oberle SG, Kanev K, Gerullis AK, Wu M, de Almeida GP, Puleston DJ, Baixauli F, Aly L, Greco A, Nizharadze T, Becker NB, Hoesslin MV, Donhauser LV, Berner J, Chu T, McNamara HA, Esencan Z, Roelli P, Wurmser C, Kleiter I, Vehreschild MJGT, Mayer CA, Knolle P, Klingenspor M, Fumagalli V, Iannacone M, Prlic M, Korn T, Pearce EL, Höfer T, Schulz AM, and Zehn D
- Subjects
- Cell Cycle, Cell Differentiation, Pyrimidines
- Abstract
Blocking pyrimidine de novo synthesis by inhibiting dihydroorotate dehydrogenase is used to treat autoimmunity and prevent expansion of rapidly dividing cell populations including activated T cells. Here we show memory T cell precursors are resistant to pyrimidine starvation. Although the treatment effectively blocked effector T cells, the number, function and transcriptional profile of memory T cells and their precursors were unaffected. This effect occurred in a narrow time window in the early T cell expansion phase when developing effector, but not memory precursor, T cells are vulnerable to pyrimidine starvation. This vulnerability stems from a higher proliferative rate of early effector T cells as well as lower pyrimidine synthesis capacity when compared with memory precursors. This differential sensitivity is a drug-targetable checkpoint that efficiently diminishes effector T cells without affecting the memory compartment. This cell fate checkpoint might therefore lead to new methods to safely manipulate effector T cell responses., (© 2023. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
33. Functional T cells are capable of supernumerary cell division and longevity.
- Author
-
Soerens AG, Künzli M, Quarnstrom CF, Scott MC, Swanson L, Locquiao JJ, Ghoneim HE, Zehn D, Youngblood B, Vezys V, and Masopust D
- Subjects
- Animals, Mice, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes immunology, Cell Differentiation, Immunologic Memory, Neoplasms immunology, Neoplasms pathology, Immunization, Secondary, Vaccination, Adoptive Transfer, Time Factors, Infections immunology, Chronic Disease, Epigenesis, Genetic, Cell Division, Longevity immunology, T-Lymphocytes cytology, T-Lymphocytes immunology, Cellular Senescence immunology, Cellular Senescence physiology, Lymphocyte Activation
- Abstract
Differentiated somatic mammalian cells putatively exhibit species-specific division limits that impede cancer but may constrain lifespans
1-3 . To provide immunity, transiently stimulated CD8+ T cells undergo unusually rapid bursts of numerous cell divisions, and then form quiescent long-lived memory cells that remain poised to reproliferate following subsequent immunological challenges. Here we addressed whether T cells are intrinsically constrained by chronological or cell-division limits. We activated mouse T cells in vivo using acute heterologous prime-boost-boost vaccinations4 , transferred expanded cells to new mice, and then repeated this process iteratively. Over 10 years (greatly exceeding the mouse lifespan)5 and 51 successive immunizations, T cells remained competent to respond to vaccination. Cells required sufficient rest between stimulation events. Despite demonstrating the potential to expand the starting population at least 1040 -fold, cells did not show loss of proliferation control and results were not due to contamination with young cells. Persistent stimulation by chronic infections or cancer can cause T cell proliferative senescence, functional exhaustion and death6 . We found that although iterative acute stimulations also induced sustained expression and epigenetic remodelling of common exhaustion markers (including PD1, which is also known as PDCD1, and TOX) in the cells, they could still proliferate, execute antimicrobial functions and form quiescent memory cells. These observations provide a model to better understand memory cell differentiation, exhaustion, cancer and ageing, and show that functionally competent T cells can retain the potential for extraordinary population expansion and longevity well beyond their organismal lifespan., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2023
- Full Text
- View/download PDF
34. Tapping the keg of discovery to advance T cell therapy.
- Author
-
Schulz AM, Zebley CC, Youngblood B, and Zehn D
- Subjects
- Ubiquitin-Protein Ligases, Cell- and Tissue-Based Therapy
- Published
- 2023
- Full Text
- View/download PDF
35. Secondary infections rejuvenate the intestinal CD103 + tissue-resident memory T cell pool.
- Author
-
von Hoesslin M, Kuhlmann M, de Almeida GP, Kanev K, Wurmser C, Gerullis AK, Roelli P, Berner J, and Zehn D
- Subjects
- Mice, Animals, Reinfection, CD8-Positive T-Lymphocytes, Memory T Cells, Inflammation, Immunologic Memory, Coinfection
- Abstract
Resident T lymphocytes (T
RM ) protect tissues during pathogen reexposure. Although TRM phenotype and restricted migratory pattern are established, we have a limited understanding of their response kinetics, stability, and turnover during reinfections. Such characterizations have been restricted by the absence of in vivo fate-mapping systems. We generated two mouse models, one to stably mark CD103+ T cells (a marker of TRM cells) and the other to specifically deplete CD103- T cells. Using these models, we observed that intestinal CD103+ T cells became activated during viral or bacterial reinfection, remained organ-confined, and retained their original phenotype but failed to reexpand. Instead, the population was largely rejuvenated by CD103+ T cells formed de novo during reinfections. This pattern remained unchanged upon deletion of antigen-specific circulating T cells, indicating that the lack of expansion was not due to competition with circulating subsets. Thus, although intestinal CD103+ resident T cells survived long term without antigen, they lacked the ability of classical memory T cells to reexpand. This indicated that CD103+ T cell populations could not autonomously maintain themselves. Instead, their numbers were sustained during reinfection via de novo formation from CD103- precursors. Moreover, in contrast to CD103- cells, which require antigen plus inflammation for their activation, CD103+ TRM became fully activated follwing exposure to inflammation alone. Together, our data indicate that primary CD103+ resident memory T cells lack secondary expansion potential and require CD103- precursors for their long-term maintenance.- Published
- 2022
- Full Text
- View/download PDF
36. MYB orchestrates T cell exhaustion and response to checkpoint inhibition.
- Author
-
Tsui C, Kretschmer L, Rapelius S, Gabriel SS, Chisanga D, Knöpper K, Utzschneider DT, Nüssing S, Liao Y, Mason T, Torres SV, Wilcox SA, Kanev K, Jarosch S, Leube J, Nutt SL, Zehn D, Parish IA, Kastenmüller W, Shi W, Buchholz VR, and Kallies A
- Subjects
- Cell Proliferation, Cell Self Renewal, Hepatocyte Nuclear Factor 1-alpha metabolism, Immunotherapy, L-Selectin metabolism, Precursor Cells, T-Lymphoid cytology, Precursor Cells, T-Lymphoid immunology, Viruses immunology, CD8-Positive T-Lymphocytes cytology, CD8-Positive T-Lymphocytes immunology, Programmed Cell Death 1 Receptor immunology, Programmed Cell Death 1 Receptor metabolism, Proto-Oncogene Proteins c-myb metabolism
- Abstract
CD8
+ T cells that respond to chronic viral infections or cancer are characterized by the expression of inhibitory receptors such as programmed cell death protein 1 (PD-1) and by the impaired production of cytokines. This state of restrained functionality-which is referred to as T cell exhaustion1,2 -is maintained by precursors of exhausted T (TPEX ) cells that express the transcription factor T cell factor 1 (TCF1), self-renew and give rise to TCF1- exhausted effector T cells3-6 . Here we show that the long-term proliferative potential, multipotency and repopulation capacity of exhausted T cells during chronic infection are selectively preserved in a small population of transcriptionally distinct CD62L+ TPEX cells. The transcription factor MYB is not only essential for the development of CD62L+ TPEX cells and maintenance of the antiviral CD8+ T cell response, but also induces functional exhaustion and thereby prevents lethal immunopathology. Furthermore, the proliferative burst in response to PD-1 checkpoint inhibition originates exclusively from CD62L+ TPEX cells and depends on MYB. Our findings identify CD62L+ TPEX cells as a stem-like population that is central to the maintenance of long-term antiviral immunity and responsiveness to immunotherapy. Moreover, they show that MYB is a transcriptional orchestrator of two fundamental aspects of exhausted T cell responses: the downregulation of effector function and the long-term preservation of self-renewal capacity., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
37. 'Stem-like' precursors are the fount to sustain persistent CD8 + T cell responses.
- Author
-
Zehn D, Thimme R, Lugli E, de Almeida GP, and Oxenius A
- Subjects
- Cell Differentiation, CD8-Positive T-Lymphocytes, Immunologic Memory
- Abstract
Virus-specific CD8
+ T cells that differentiate in the context of resolved versus persisting infections exhibit divergent phenotypic and functional characteristics, which suggests that their differentiation trajectories are governed by distinct cellular dynamics, developmental pathways and molecular mechanisms. For acute infection, it is long known that antigen-specific T cell populations contain terminally differentiated effector T cells, known as short-lived effector T cells, and proliferation-competent and differentiation-competent memory precursor T cells. More recently, it was identified that a similar functional segregation occurs in chronic infections. A failure to generate proliferation-competent precursor cells in chronic infections and tumors results in the collapse of the T cell response. Thus, these precursor cells are major therapeutic and prophylactic targets of immune interventions. These observations suggest substantial commonality between T cell responses in acute and chronic infections but there are also critical differences. We are therefore reviewing the common features and peculiarities of precursor cells in acute infections, different types of persistent infection and cancer., (© 2022. Springer Nature America, Inc.)- Published
- 2022
- Full Text
- View/download PDF
38. T cell receptor and IL-2 signaling strength control memory CD8 + T cell functional fitness via chromatin remodeling.
- Author
-
Chin SS, Guillen E, Chorro L, Achar S, Ng K, Oberle S, Alfei F, Zehn D, Altan-Bonnet G, Delahaye F, and Lauvau G
- Subjects
- Antigens metabolism, CD8-Positive T-Lymphocytes, Calcium metabolism, Chromatin metabolism, Chromatin Assembly and Disassembly, Immunologic Memory, Receptors, Antigen, T-Cell metabolism, Biological Phenomena, Interleukin-2
- Abstract
Cognate antigen signal controls CD8
+ T cell priming, expansion size and effector versus memory cell fates, but it is not known if and how it modulates the functional features of memory CD8+ T cells. Here we show that the strength of T cell receptor (TCR) signaling controls the requirement for interleukin-2 (IL-2) signals to form a pool of memory CD8+ T cells that competitively re-expand upon secondary antigen encounter. Combining strong TCR and intact IL-2 signaling during priming synergistically induces genome-wide chromatin accessibility in regions targeting a wide breadth of biological processes, consistent with greater T cell functional fitness. Chromatin accessibility in promoters of genes encoding for stem cell, cell cycle and calcium-related proteins correlates with faster intracellular calcium accumulation, initiation of cell cycle and more robust expansion. High-dimensional flow-cytometry analysis of these T cells also highlights higher diversity of T cell subsets and phenotypes with T cells primed with stronger TCR and IL-2 stimulation than those primed with weaker strengths of TCR and/or IL-2 signals. These results formally show that epitope selection in vaccine design impacts memory CD8+ T cell epigenetic programming and function., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
39. Type 1 conventional dendritic cells maintain and guide the differentiation of precursors of exhausted T cells in distinct cellular niches.
- Author
-
Dähling S, Mansilla AM, Knöpper K, Grafen A, Utzschneider DT, Ugur M, Whitney PG, Bachem A, Arampatzi P, Imdahl F, Kaisho T, Zehn D, Klauschen F, Garbi N, Kallies A, Saliba AE, Gasteiger G, Bedoui S, and Kastenmüller W
- Subjects
- Cell Differentiation, Immunotherapy, Lymphocyte Count, CD8-Positive T-Lymphocytes, Dendritic Cells
- Abstract
Reinvigoration of exhausted CD8
+ T (Tex) cells by checkpoint immunotherapy depends on the activation of precursors of exhausted T (Tpex) cells, but the local anatomical context of their maintenance, differentiation, and interplay with other cells is not well understood. Here, we identified transcriptionally distinct Tpex subpopulations, mapped their differentiation trajectories via transitory cellular states toward Tex cells, and localized these cell states to specific splenic niches. Conventional dendritic cells (cDCs) were critical for successful αPD-L1 therapy and were required to mediate viral control. cDC1s were dispensable for Tpex cell expansion but provided an essential niche to promote Tpex cell maintenance, preventing their overactivation and T-cell-mediated immunopathology. Mechanistically, cDC1s insulated Tpex cells via MHC-I-dependent interactions to prevent their activation within other inflammatory environments that further aggravated their exhaustion. Our findings reveal that cDC1s maintain and safeguard Tpex cells within distinct anatomical niches to balance viral control, exhaustion, and immunopathology., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
40. Mastering an exhausting marathon: how CD8 + T cells fine-tune metabolic fitness.
- Author
-
Schulz AM and Zehn D
- Subjects
- Cell Differentiation, Lymphocyte Activation, CD8-Positive T-Lymphocytes
- Abstract
A recent study by Gabriel et al. provides novel insight into the metabolic pathways that contribute to T cell differentiation in chronic infection. The researchers discovered that metabolic plasticity and the function of exhausted T cells is regulated via the TGF-β-mTOR signaling axis., (© 2022 The Authors. Immunology & Cell Biology published by John Wiley & Sons Australia, Ltd on behalf of Australian and New Zealand Society for Immunology, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
41. Dynamics of spike-and nucleocapsid specific immunity during long-term follow-up and vaccination of SARS-CoV-2 convalescents.
- Author
-
Koerber N, Priller A, Yazici S, Bauer T, Cheng CC, Mijočević H, Wintersteller H, Jeske S, Vogel E, Feuerherd M, Tinnefeld K, Winter C, Ruland J, Gerhard M, Haller B, Christa C, Zelger O, Roggendorf H, Halle M, Erber J, Lingor P, Keppler O, Zehn D, Protzer U, and Knolle PA
- Subjects
- Antibodies, Neutralizing immunology, Antibodies, Viral immunology, BNT162 Vaccine administration & dosage, COVID-19 virology, Cytokines immunology, Cytokines metabolism, Flow Cytometry methods, Follow-Up Studies, Humans, Immunoglobulin G immunology, Interleukin-2 immunology, Interleukin-2 metabolism, Kinetics, SARS-CoV-2 physiology, T-Lymphocytes immunology, T-Lymphocytes metabolism, T-Lymphocytes virology, Time Factors, Vaccination methods, BNT162 Vaccine immunology, COVID-19 immunology, Convalescence, Nucleocapsid immunology, SARS-CoV-2 immunology, Spike Glycoprotein, Coronavirus immunology
- Abstract
Anti-viral immunity continuously declines over time after SARS-CoV-2 infection. Here, we characterize the dynamics of anti-viral immunity during long-term follow-up and after BNT162b2 mRNA-vaccination in convalescents after asymptomatic or mild SARS-CoV-2 infection. Virus-specific and virus-neutralizing antibody titers rapidly declined in convalescents over 9 months after infection, whereas virus-specific cytokine-producing polyfunctional T cells persisted, among which IL-2-producing T cells correlated with virus-neutralizing antibody titers. Among convalescents, 5% of individuals failed to mount long-lasting immunity after infection and showed a delayed response to vaccination compared to 1% of naïve vaccinees, but successfully responded to prime/boost vaccination. During the follow-up period, 8% of convalescents showed a selective increase in virus-neutralizing antibody titers without accompanying increased frequencies of circulating SARS-CoV-2-specific T cells. The same convalescents, however, responded to vaccination with simultaneous increase in antibody and T cell immunity revealing the strength of mRNA-vaccination to increase virus-specific immunity in convalescents., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
42. Siglec-H-Deficient Mice Show Enhanced Type I IFN Responses, but Do Not Develop Autoimmunity After Influenza or LCMV Infections.
- Author
-
Szumilas N, Corneth OBJ, Lehmann CHK, Schmitt H, Cunz S, Cullen JG, Chu T, Marosan A, Mócsai A, Benes V, Zehn D, Dudziak D, Hendriks RW, and Nitschke L
- Subjects
- Animals, Autoimmune Diseases virology, Autoimmunity immunology, Chemotaxis, Leukocyte immunology, Dendritic Cells immunology, Influenza A Virus, H3N2 Subtype, Lectins deficiency, Lymphocytic choriomeningitis virus, Mice, Mice, Inbred C57BL, Mice, Knockout, Receptors, Cell Surface deficiency, Toll-Like Receptor 9 immunology, Toll-Like Receptor 9 metabolism, Arenaviridae Infections immunology, Autoimmune Diseases immunology, Interferon Type I immunology, Lectins immunology, Orthomyxoviridae Infections immunology, Receptors, Cell Surface immunology
- Abstract
Siglec-H is a DAP12-associated receptor on plasmacytoid dendritic cells (pDCs) and microglia. Siglec-H inhibits TLR9-induced IFN-α production by pDCs. Previously, it was found that Siglec-H-deficient mice develop a lupus-like severe autoimmune disease after persistent murine cytomegalovirus (mCMV) infection. This was due to enhanced type I interferon responses, including IFN-α. Here we examined, whether other virus infections can also induce autoimmunity in Siglec-H-deficient mice. To this end we infected Siglec-H-deficient mice with influenza virus or with Lymphocytic Choriomeningitis virus (LCMV) clone 13. With both types of viruses we did not observe induction of autoimmune disease in Siglec-H-deficient mice. This can be explained by the fact that both types of viruses are ssRNA viruses that engage TLR7, rather than TLR9. Also, Influenza causes an acute infection that is rapidly cleared and the chronicity of LCMV clone 13 may not be sufficient and may rather suppress pDC functions. Siglec-H inhibited exclusively TLR-9 driven type I interferon responses, but did not affect type II or type III interferon production by pDCs. Siglec-H-deficient pDCs showed impaired Hck expression, which is a Src-family kinase expressed in myeloid cells, and downmodulation of the chemokine receptor CCR9, that has important functions for pDCs. Accordingly, Siglec-H-deficient pDCs showed impaired migration towards the CCR9 ligand CCL25. Furthermore, autoimmune-related genes such as Klk1 and DNase1l3 are downregulated in Siglec-H-deficient pDCs as well. From these findings we conclude that Siglec-H controls TLR-9-dependent, but not TLR-7 dependent inflammatory responses after virus infections and regulates chemokine responsiveness of pDCs., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Szumilas, Corneth, Lehmann, Schmitt, Cunz, Cullen, Chu, Marosan, Mócsai, Benes, Zehn, Dudziak, Hendriks and Nitschke.)
- Published
- 2021
- Full Text
- View/download PDF
43. The role of exhaustion in CAR T cell therapy.
- Author
-
Delgoffe GM, Xu C, Mackall CL, Green MR, Gottschalk S, Speiser DE, Zehn D, and Beavis PA
- Subjects
- Humans, Immunotherapy, Adoptive methods, Neoplasms immunology, Tumor Microenvironment, Drug Resistance, Neoplasm, Immunotherapy, Adoptive standards, Lymphocytes, Tumor-Infiltrating immunology, Neoplasms therapy, Receptors, Chimeric Antigen immunology
- Abstract
CAR T cell therapy successes are challenged by several mechanisms of resistance including the development of dysfunctional states such as exhaustion. The features of CAR T cell exhaustion, its role in limiting the efficacy of CAR T therapy in both liquid and solid malignancies, and potential strategies to overcome it are discussed., (Crown Copyright © 2021. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
44. Inflammatory signals are sufficient to elicit TOX expression in mouse and human CD8+ T cells.
- Author
-
Maurice NJ, Berner J, Taber AK, Zehn D, and Prlic M
- Subjects
- Animals, Cytokines immunology, Humans, Immunologic Memory, Lymphocyte Activation, Mice, Programmed Cell Death 1 Receptor metabolism, Receptors, Antigen, T-Cell, Signal Transduction, CD8-Positive T-Lymphocytes immunology, Gene Expression Regulation immunology, High Mobility Group Proteins metabolism, Homeodomain Proteins metabolism, Inflammation metabolism
- Abstract
T cell receptor (TCR) stimulation leads to the expression of the transcription factor thymocyte selection-associated high-mobility group box (TOX). Prolonged TCR signaling, such as encountered during chronic infections or in tumors, leads to sustained TOX expression, which is required for the induction of a state of exhaustion or dysfunction. Although CD8+ memory T (Tmem) cells in mice typically do not express TOX at steady state, some human Tmem cells express TOX but appear fully functional. This seeming discrepancy between mouse and human T cells has led to the speculation that TOX is differentially regulated between these species, which could complicate the interpretation of preclinical mouse model studies. We report here that, similar to TCR-mediated signals, inflammatory cytokines are also sufficient to increase TOX expression in human and mouse Tmem cells. Thus, TOX expression is controlled by the environment, which provides an explanation for the different TOX expression patterns encountered in T cells isolated from specific pathogen-free laboratory mice versus humans. Finally, we report that TOX is not necessary for cytokine-driven expression of programmed cell death 1. Overall, our data highlight that the mechanisms regulating TOX expression are conserved across species and indicate that TOX expression reflects a T cell's activation state and does not necessarily correlate with T cell dysfunction.
- Published
- 2021
- Full Text
- View/download PDF
45. Cervicovaginal Tissue Residence Confers a Distinct Differentiation Program upon Memory CD8 T Cells.
- Author
-
Davé VA, Cardozo-Ojeda EF, Mair F, Erickson J, Woodward-Davis AS, Koehne A, Soerens A, Czartoski J, Teague C, Potchen N, Oberle S, Zehn D, Schiffer JT, Lund JM, and Prlic M
- Subjects
- Adult, Animals, Cell Differentiation drug effects, Cell Differentiation immunology, Cervix Uteri drug effects, Cervix Uteri virology, Female, Herpes Simplex drug therapy, Herpes Simplex virology, Herpesvirus 2, Human drug effects, Herpesvirus 2, Human immunology, Humans, Injections, Subcutaneous, Medroxyprogesterone Acetate administration & dosage, Medroxyprogesterone Acetate pharmacology, Mice, Mice, Inbred C57BL, Vagina drug effects, Vagina virology, Young Adult, CD8-Positive T-Lymphocytes immunology, Cervix Uteri immunology, Herpes Simplex immunology, Vagina immunology
- Abstract
Tissue-resident memory CD8 T cells (CD8 T
RM ) are critical for maintaining barrier immunity. CD8 TRM have been mainly studied in the skin, lung and gut, with recent studies suggesting that the signals that control tissue residence and phenotype are highly tissue dependent. We examined the T cell compartment in healthy human cervicovaginal tissue (CVT) and found that most CD8 T cells were granzyme B+ and TCF-1- To address if this phenotype is driven by CVT tissue residence, we used a mouse model to control for environmental factors. Using localized and systemic infection models, we found that CD8 TRM in the mouse CVT gradually acquired a granzyme B+ , TCF-1- phenotype as seen in human CVT. In contrast to CD8 TRM in the gut, these CD8 TRM were not stably maintained regardless of the initial infection route, which led to reductions in local immunity. Our data show that residence in the CVT is sufficient to progressively shape the size and function of its CD8 TRM compartment., (Copyright © 2021 by The American Association of Immunologists, Inc.)- Published
- 2021
- Full Text
- View/download PDF
46. Author Correction: Auto-aggressive CXCR6 + CD8 T cells cause liver immune pathology in NASH.
- Author
-
Dudek M, Pfister D, Donakonda S, Filpe P, Schneider A, Laschinger M, Hartmann D, Hüser N, Meiser P, Bayerl F, Inverso D, Wigger J, Sebode M, Öllinger R, Rad R, Hegenbarth S, Anton M, Guillot A, Bowman A, Heide D, Müller F, Ramadori P, Leone V, Garcia-Caceres C, Gruber T, Seifert G, Kabat AM, Mallm JP, Reider S, Effenberger M, Roth S, Billeter AT, Müller-Stich B, Pearce EJ, Koch-Nolte F, Käser R, Tilg H, Thimme R, Boettler T, Tacke F, Dufour JF, Haller D, Murray PJ, Heeren R, Zehn D, Böttcher JP, Heikenwälder M, and Knolle PA
- Published
- 2021
- Full Text
- View/download PDF
47. Auto-aggressive CXCR6 + CD8 T cells cause liver immune pathology in NASH.
- Author
-
Dudek M, Pfister D, Donakonda S, Filpe P, Schneider A, Laschinger M, Hartmann D, Hüser N, Meiser P, Bayerl F, Inverso D, Wigger J, Sebode M, Öllinger R, Rad R, Hegenbarth S, Anton M, Guillot A, Bowman A, Heide D, Müller F, Ramadori P, Leone V, Garcia-Caceres C, Gruber T, Seifert G, Kabat AM, Mallm JP, Reider S, Effenberger M, Roth S, Billeter AT, Müller-Stich B, Pearce EJ, Koch-Nolte F, Käser R, Tilg H, Thimme R, Boettler T, Tacke F, Dufour JF, Haller D, Murray PJ, Heeren R, Zehn D, Böttcher JP, Heikenwälder M, and Knolle PA
- Subjects
- Acetates pharmacology, Animals, CD8-Positive T-Lymphocytes drug effects, CD8-Positive T-Lymphocytes pathology, Cell Death drug effects, Cell Death immunology, Diet, High-Fat adverse effects, Disease Models, Animal, Humans, Interleukin-15 immunology, Interleukin-15 pharmacology, Liver drug effects, Male, Mice, Mice, Inbred C57BL, CD8-Positive T-Lymphocytes immunology, Liver immunology, Liver pathology, Non-alcoholic Fatty Liver Disease immunology, Non-alcoholic Fatty Liver Disease pathology, Receptors, CXCR6 immunology
- Abstract
Nonalcoholic steatohepatitis (NASH) is a manifestation of systemic metabolic disease related to obesity, and causes liver disease and cancer
1,2 . The accumulation of metabolites leads to cell stress and inflammation in the liver3 , but mechanistic understandings of liver damage in NASH are incomplete. Here, using a preclinical mouse model that displays key features of human NASH (hereafter, NASH mice), we found an indispensable role for T cells in liver immunopathology. We detected the hepatic accumulation of CD8 T cells with phenotypes that combined tissue residency (CXCR6) with effector (granzyme) and exhaustion (PD1) characteristics. Liver CXCR6+ CD8 T cells were characterized by low activity of the FOXO1 transcription factor, and were abundant in NASH mice and in patients with NASH. Mechanistically, IL-15 induced FOXO1 downregulation and CXCR6 upregulation, which together rendered liver-resident CXCR6+ CD8 T cells susceptible to metabolic stimuli (including acetate and extracellular ATP) and collectively triggered auto-aggression. CXCR6+ CD8 T cells from the livers of NASH mice or of patients with NASH had similar transcriptional signatures, and showed auto-aggressive killing of cells in an MHC-class-I-independent fashion after signalling through P2X7 purinergic receptors. This killing by auto-aggressive CD8 T cells fundamentally differed from that by antigen-specific cells, which mechanistically distinguishes auto-aggressive and protective T cell immunity.- Published
- 2021
- Full Text
- View/download PDF
48. Loss of the orphan nuclear receptor NR2F6 enhances CD8 + T-cell memory via IFN-γ.
- Author
-
Jakic B, Olson WJ, Siegmund K, Klepsch V, Kimpel J, Labi V, Zehn D, Baier G, and Hermann-Kleiter N
- Subjects
- Animals, Female, Male, Mice, Mice, Inbred C57BL, Repressor Proteins deficiency, CD8-Positive T-Lymphocytes immunology, Interferon-gamma immunology, Orphan Nuclear Receptors immunology, Repressor Proteins immunology
- Abstract
Memory formation is a hallmark of T cell-mediated immunity, but how differentiation into either short-lived effector cells (SLECs, CD127
- KLRG1+ ) or memory precursors cells (MPECs, CD127+ KLRG1- ) and subsequent regulation of long-term memory is adjusted is incompletely understood. Here, we show that loss of the nuclear orphan receptor NR2F6 in germ-line Nr2f6-deficient mice enhances antigen-specific CD8+ memory formation up to 70 days after bacterial infection with Listeria monocytogenes (LmOVA) and boosts inflammatory IFN-γ, TNFα, and IL-2 cytokine recall responses. Adoptive transfer experiments using Nr2f6-/- OT-I T-cells showed that the augmented memory formation is CD8+ T-cell intrinsic. Although the relative difference between the Nr2f6+/+ and Nr2f6-/- OT-I memory compartment declines over time, Nr2f6-deficient OT-I memory T cells mount significantly enhanced IFN-γ responses upon reinfection with increased clonal expansion and improved host antigen-specific CD8+ T-cell responses. Following a secondary adoptive transfer into naïve congenic mice, Nr2f6-deficient OT-I memory T cells are superior in clearing LmOVA infection. Finally, we show that the commitment to enhanced memory within Nr2f6-deficient OT-I T cells is established in the early phases of the antibacterial immune response and is IFN-γ mediated. IFN-γ blocking normalized MPEC formation of Nr2f6-deficient OT-I T cells. Thus, deletion or pharmacological inhibition of NR2F6 in antigen-specific CD8+ T cells may have therapeutic potential for enhancing early IFN-γ production and consequently the functionality of memory CD8+ T cells in vivo.- Published
- 2021
- Full Text
- View/download PDF
49. Enabling single-cell trajectory network enrichment.
- Author
-
Grønning AGB, Oubounyt M, Kanev K, Lund J, Kacprowski T, Zehn D, Röttger R, and Baumbach J
- Abstract
Single-cell sequencing (scRNA-seq) technologies allow the investigation of cellular differentiation processes with unprecedented resolution. Although powerful software packages for scRNA-seq data analysis exist, systems biology-based tools for trajectory analysis are rare and typically difficult to handle. This hampers biological exploration and prevents researchers from gaining deeper insights into the molecular control of developmental processes. Here, to address this, we have developed Scellnetor; a network-constraint time-series clustering algorithm. It allows extraction of temporal differential gene expression network patterns (modules) that explain the difference in regulation of two developmental trajectories. Using well-characterized experimental model systems, we demonstrate the capacity of Scellnetor as a hypothesis generator to identify putative mechanisms driving haematopoiesis or mechanistically interpretable subnetworks driving dysfunctional CD8 T-cell development in chronic infections. Altogether, Scellnetor allows for single-cell trajectory network enrichment, which effectively lifts scRNA-seq data analysis to a systems biology level., (© 2021. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2021
- Full Text
- View/download PDF
50. Origin and fine-tuning of effector CD8 T cell subpopulations in chronic infection.
- Author
-
Kanev K and Zehn D
- Subjects
- Animals, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes metabolism, Chronic Disease, Epigenesis, Genetic, High Mobility Group Proteins genetics, Homeodomain Proteins genetics, Humans, Immunologic Memory, Mice, Precursor Cells, T-Lymphoid immunology, Precursor Cells, T-Lymphoid metabolism, T-Lymphocyte Subsets metabolism, T-Lymphocytes, Helper-Inducer immunology, Transcription, Genetic, CD8-Positive T-Lymphocytes immunology, High Mobility Group Proteins metabolism, Homeodomain Proteins metabolism, T-Lymphocyte Subsets immunology, Virus Diseases immunology
- Abstract
Persisting stimulation can skew CD8 T cells towards a hypofunctional state commonly referred to as T cell exhaustion. This functional attenuation likely constitutes a mechanism which evolved to balance T cell mediated viral control versus overwhelming immunopathology. Here, we highlight the recent progress in defining the genetic mechanisms and factors shaping the differentiation of exhausted CD8 T cells. We review how the transcription factor Tox imposes an exhausted phenotype in the Tcf1+ progenitors and how CD4 help fine-tunes the effector subsets that emerge from this progenitor population. Both processes critically shape the spectrum of effector function performed by CD8 T cells and the level of resulting virus control. Finally, we discuss how these insights can be exploited to boost the immune response in chronic infection and cancer., (Copyright © 2020 Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.