1
|
Cardani-Boulton A, Lin F, Bergmann CC. CD6 regulates CD4 T follicular helper cell differentiation and humoral immunity during murine coronavirus infection. J Virol 2025; 99:e0186424. [PMID: 39679790 PMCID: PMC11784103 DOI: 10.1128/jvi.01864-24] [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: 10/23/2024] [Accepted: 11/25/2024] [Indexed: 12/17/2024] Open
Abstract
During activation, the T cell transmembrane receptor CD6 becomes incorporated into the T cell immunological synapse where it can exert both co-stimulatory and co-inhibitory functions. Given the ability of CD6 to carry out opposing functions, this study sought to determine how CD6 regulates early T cell activation in response to viral infection. Infection of CD6-deficient mice with a neurotropic murine coronavirus resulted in greater activation and expansion of CD4 T cells in the draining lymph nodes. Further analysis demonstrated that there was also preferential differentiation of CD4 T cells into T follicular helper cells, resulting in accelerated germinal center responses and emergence of high-affinity virus-specific antibodies. Given that CD6 conversely supports CD4 T cell activation in many autoimmune models, we probed potential mechanisms of CD6-mediated suppression of CD4 T cell activation during viral infection. Analysis of CD6 binding proteins revealed that infection-induced upregulation of Ubash3a, a negative regulator of T cell receptor (TCR) signaling, was hindered in CD6-deficient lymph nodes. Consistent with greater T cell activation and reduced UBASH3a activity, the T cell receptor signal strength was intensified in CD6-deficient CD4 T cells. These results reveal a novel immunoregulatory role for CD6 in limiting CD4 T cell activation and deterring CD4 T follicular helper cell differentiation, thereby attenuating antiviral humoral immunity. IMPORTANCE CD6 monoclonal blocking antibodies are being therapeutically administered to inhibit T cell activation in autoimmune disorders. However, the multifaceted nature of CD6 allows for multiple and even opposing functions under different circumstances of T cell activation. We therefore sought to characterize how CD6 regulates T cell activation in the context of viral infections using an in vivo murine coronavirus model. In contrast to its role in autoimmunity, but consistent with its function in the presence of superantigens, we found that CD6 deficiency enhances CD4 T cell activation and CD4 T cell help to germinal center-dependent antiviral humoral responses. Finally, we provide evidence that CD6 regulates transcription of its intracellular binding partner UBASH3a, which suppresses T cell receptor (TCR) signaling and consequently T cell activation. These findings highlight the context-dependent flexibility of CD6 in regulating in vivo adaptive immune responses, which may be targeted to enhance antiviral immunity.
Collapse
MESH Headings
- Animals
- Mice
- Immunity, Humoral/immunology
- Cell Differentiation/immunology
- T Follicular Helper Cells/immunology
- Lymphocyte Activation/immunology
- Antigens, Differentiation, T-Lymphocyte/genetics
- Antigens, Differentiation, T-Lymphocyte/immunology
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Coronavirus Infections/immunology
- Coronavirus Infections/virology
- Mice, Inbred C57BL
- Germinal Center/immunology
- Mice, Knockout
- CD4-Positive T-Lymphocytes/immunology
- Antigens, CD/immunology
- Antigens, CD/genetics
- T-Lymphocytes, Helper-Inducer/immunology
- Signal Transduction
- Murine hepatitis virus/immunology
- Antibodies, Viral/immunology
Collapse
Affiliation(s)
- Amber Cardani-Boulton
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Feng Lin
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Department of Immunity and Inflammation, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Cornelia C. Bergmann
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
2
|
Manjili MH, Manjili SH. The quantum model of T-cell activation: Revisiting immune response theories. Scand J Immunol 2024; 100:e13375. [PMID: 38750629 PMCID: PMC11250909 DOI: 10.1111/sji.13375] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 03/15/2024] [Accepted: 04/29/2024] [Indexed: 07/16/2024]
Abstract
Our understanding of the immune response is far from complete, missing out on more detailed explanations that could be provided by molecular insights. To bridge this gap, we introduce the quantum model of T-cell activation. This model suggests that the transfer of energy during protein phosphorylation within T cells is not a continuous flow but occurs in discrete bursts, or 'quanta', of phosphates. This quantized energy transfer is mediated by oscillating cycles of receptor phosphorylation and dephosphorylation, initiated by dynamic 'catch-slip' pulses in the peptide-major histocompatibility complex-T-cell receptor (pMHC-TcR) interactions. T-cell activation is predicated upon achieving a critical threshold of catch-slip pulses at the pMHC-TcR interface. Costimulation is relegated to a secondary role, becoming crucial only when the frequency of pMHC-TcR catch-slip pulses does not meet the necessary threshold for this quanta-based energy transfer. Therefore, our model posits that it is the quantum nature of energy transfer-not the traditional signal I or signal II-that plays the decisive role in T-cell activation. This paradigm shift highlights the importance of understanding T-cell activation through a quantum lens, offering a potentially transformative perspective on immune response regulation.
Collapse
Affiliation(s)
- Masoud H. Manjili
- Department of Microbiology & Immunology, VCU School of Medicine
- Massey Comprehensive Cancer Center, 401 College Street, Richmond, VA, 23298, USA
| | - Saeed H. Manjili
- AMF Automation Technologies LLC, 2115 W. Laburnum Ave., Richmond, VA 23227
| |
Collapse
|
3
|
Cardani-Boulton A, Lin F, Bergmann CC. CD6 Regulates CD4 T Follicular Helper Cell Differentiation and Humoral Immunity During Murine Coronavirus Infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.26.605237. [PMID: 39091786 PMCID: PMC11291160 DOI: 10.1101/2024.07.26.605237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
During activation the T cell transmembrane receptor CD6 becomes incorporated into the T cell immunological synapse where it can exert both co-stimulatory and co-inhibitory functions. Given the ability of CD6 to carry out opposing functions, this study sought to determine how CD6 regulates early T cell activation in response to viral infection. Infection of CD6 deficient mice with a neurotropic murine coronavirus resulted in greater activation and expansion of CD4 T cells in the draining lymph nodes. Further analysis demonstrated that there was also preferential differentiation of CD4 T cells into T follicular helper cells, resulting in accelerated germinal center responses and emergence of high affinity virus specific antibodies. Given that CD6 conversely supports CD4 T cell activation in many autoimmune models, we probed potential mechanisms of CD6 mediated suppression of CD4 T cell activation during viral infection. Analysis of CD6 binding proteins revealed that infection induced upregulation of Ubash3a, a negative regulator of T cell receptor signaling, was hindered in CD6 deficient lymph nodes. Consistent with greater T cell activation and reduced UBASH3a activity, the T cell receptor signal strength was intensified in CD6 deficient CD4 T cells. These results reveal a novel immunoregulatory role for CD6 in limiting CD4 T cell activation and deterring CD4 T follicular helper cell differentiation, thereby attenuating antiviral humoral immunity.
Collapse
Affiliation(s)
- Amber Cardani-Boulton
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Case Western Reserve University, Cleveland Clinic, Lerner College of Medicine, Cleveland, OH
| | - Feng Lin
- Case Western Reserve University, Cleveland Clinic, Lerner College of Medicine, Cleveland, OH
- Department of Immunity and Inflammation, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Cornelia C Bergmann
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Case Western Reserve University, Cleveland Clinic, Lerner College of Medicine, Cleveland, OH
| |
Collapse
|
4
|
Konecny AJ, Huang Y, Setty M, Prlic M. Signals that control MAIT cell function in healthy and inflamed human tissues. Immunol Rev 2024; 323:138-149. [PMID: 38520075 PMCID: PMC12045158 DOI: 10.1111/imr.13325] [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] [Indexed: 03/25/2024]
Abstract
Mucosal-associated invariant T (MAIT) cells have a semi-invariant T-cell receptor that allows recognition of antigen in the context of the MHC class I-related (MR1) protein. Metabolic intermediates of the riboflavin synthesis pathway have been identified as MR1-restricted antigens with agonist properties. As riboflavin synthesis occurs in many bacterial species, but not human cells, it has been proposed that the main purpose of MAIT cells is antibacterial surveillance and protection. The majority of human MAIT cells secrete interferon-gamma (IFNg) upon activation, while some MAIT cells in tissues can also express IL-17. Given that MAIT cells are present in human barrier tissues colonized by a microbiome, MAIT cells must somehow be able to distinguish colonization from infection to ensure effector functions are only elicited when necessary. Importantly, MAIT cells have additional functional properties, including the potential to contribute to restoring tissue homeostasis by expression of CTLA-4 and secretion of the cytokine IL-22. A recent study provided compelling data indicating that the range of human MAIT cell functional properties is explained by plasticity rather than distinct lineages. This further underscores the necessity to better understand how different signals regulate MAIT cell function. In this review, we highlight what is known in regards to activating and inhibitory signals for MAIT cells with a specific focus on signals relevant to healthy and inflamed tissues. We consider the quantity, quality, and the temporal order of these signals on MAIT cell function and discuss the current limitations of computational tools to extrapolate which signals are received by MAIT cells in human tissues. Using lessons learned from conventional CD8 T cells, we also discuss how TCR signals may integrate with cytokine signals in MAIT cells to elicit distinct functional states.
Collapse
Affiliation(s)
- Andrew J. Konecny
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Immunology, University of Washington, Seattle, Washington, USA
| | - Yin Huang
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Herbold Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, Washington, USA
| | - Manu Setty
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Herbold Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Martin Prlic
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Immunology, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
| |
Collapse
|
5
|
Min H, Valente LA, Xu L, O'Neil SM, Begg LR, Kurtzberg J, Filiano AJ. Improving thymus implantation for congenital athymia with interleukin-7. Clin Transl Immunology 2023; 12:e1475. [PMID: 38020730 PMCID: PMC10665642 DOI: 10.1002/cti2.1475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 11/03/2023] [Accepted: 11/08/2023] [Indexed: 12/01/2023] Open
Abstract
Objectives Thymus implantation is a recently FDA-approved therapy for congenital athymia. Patients receiving thymus implantation develop a functional but incomplete T cell compartment. Our objective was to develop a mouse model to study clinical thymus implantation in congenital athymia and to optimise implantation procedures to maximise T cell education and expansion of naïve T cells. Methods Using Foxn1 nu athymic mice as recipients, we tested MHC-matched and -mismatched donor thymi that were implanted as fresh tissue or cultured to remove donor T cells. We first implanted thymus under the kidney capsule and then optimised intramuscular implantation. Using competitive adoptive transfer assays, we investigated whether the failure of newly developed T cells to expand into a complete T cell compartment was because of intrinsic deficits or whether there were deficits in engaging MHC molecules in the periphery. Finally, we tested whether recombinant IL-7 would promote the expansion of host naïve T cells educated by the implanted thymus. Results We determined that thymus implants in Foxn1 nu athymic mice mimic many aspects of clinical thymus implants in patients with congenital athymia. When we implanted cultured, MHC-mismatched donor thymus into Foxn1 nu athymic mice, mice developed a limited T cell compartment with notably underdeveloped naïve populations and overrepresented memory-like T cells. Newly generated T cells were predominantly educated by MHC molecules expressed by the donor thymus, thus potentially undergoing another round of selection once in the peripheral circulation. Using competitive adoptive transfer assays, we compared expansion rates of T cells educated on donor thymus versus T cells educated during typical thymopoiesis in MHC-matched and -mismatched environments. Once in the circulation, regardless of the MHC haplotypes, T cells educated on a donor thymus underwent abnormal expansion with initially more robust proliferation coupled with greater cell death, resembling IL-7 independent spontaneous expansion. Treating implanted mice with recombinant interleukin (IL-7) promoted homeostatic expansion that improved T cell development, expanded the T cell receptor repertoire, and normalised the naïve T cell compartment. Conclusion We conclude that implanting cultured thymus into the muscle of Foxn1 nu athymic mice is an appropriate system to study thymus implantation for congenital athymia and immunodeficiencies. T cells are educated by the donor thymus, yet naïve T cells have deficits in expansion. IL-7 greatly improves T cell development after thymus implantation and may offer a novel strategy to improve outcomes of clinical thymus implantation.
Collapse
Affiliation(s)
- Hyunjung Min
- Marcus Center for Cellular CuresDuke UniversityDurhamNCUSA
| | - Laura A Valente
- Marcus Center for Cellular CuresDuke UniversityDurhamNCUSA
- Department of PathologyDuke UniversityDurhamNCUSA
| | - Li Xu
- Marcus Center for Cellular CuresDuke UniversityDurhamNCUSA
| | - Shane M O'Neil
- Marcus Center for Cellular CuresDuke UniversityDurhamNCUSA
| | - Lauren R Begg
- Marcus Center for Cellular CuresDuke UniversityDurhamNCUSA
| | - Joanne Kurtzberg
- Marcus Center for Cellular CuresDuke UniversityDurhamNCUSA
- Department of PediatricsDuke UniversityDurhamNCUSA
| | - Anthony J Filiano
- Marcus Center for Cellular CuresDuke UniversityDurhamNCUSA
- Department of PathologyDuke UniversityDurhamNCUSA
- Department of NeurosurgeryDuke UniversityDurhamNCUSA
- Department of ImmunologyDuke UniversityDurhamNCUSA
| |
Collapse
|
6
|
Gouttefangeas C, Klein R, Maia A. The good and the bad of T cell cross-reactivity: challenges and opportunities for novel therapeutics in autoimmunity and cancer. Front Immunol 2023; 14:1212546. [PMID: 37409132 PMCID: PMC10319254 DOI: 10.3389/fimmu.2023.1212546] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 05/24/2023] [Indexed: 07/07/2023] Open
Abstract
T cells are main actors of the immune system with an essential role in protection against pathogens and cancer. The molecular key event involved in this absolutely central task is the interaction of membrane-bound specific T cell receptors with peptide-MHC complexes which initiates T cell priming, activation and recall, and thus controls a range of downstream functions. While textbooks teach us that the repertoire of mature T cells is highly diverse, it is clear that this diversity cannot possibly cover all potential foreign peptides that might be encountered during life. TCR cross-reactivity, i.e. the ability of a single TCR to recognise different peptides, offers the best solution to this biological challenge. Reports have shown that indeed, TCR cross-reactivity is surprisingly high. Hence, the T cell dilemma is the following: be as specific as possible to target foreign danger and spare self, while being able to react to a large spectrum of body-threatening situations. This has major consequences for both autoimmune diseases and cancer, and significant implications for the development of T cell-based therapies. In this review, we will present essential experimental evidence of T cell cross-reactivity, implications for two opposite immune conditions, i.e. autoimmunity vs cancer, and how this can be differently exploited for immunotherapy approaches. Finally, we will discuss the tools available for predicting cross-reactivity and how improvements in this field might boost translational approaches.
Collapse
Affiliation(s)
- Cécile Gouttefangeas
- Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Tübingen, Germany
| | - Reinhild Klein
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Ana Maia
- Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
| |
Collapse
|
7
|
Park JS, Chung IJ, Kim HR, Jun CD. The Immunosuppressive Potential of Cholesterol Sulfate Through T Cell Microvilli Disruption. Immune Netw 2023; 23:e29. [PMID: 37416932 PMCID: PMC10320417 DOI: 10.4110/in.2023.23.e29] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/28/2023] [Accepted: 04/21/2023] [Indexed: 07/08/2023] Open
Abstract
Cholesterol (CL) is required for various biomolecular production processes, including those of cell membrane components. Therefore, to meet these needs, CL is converted into various derivatives. Among these derivatives is cholesterol sulfate (CS), a naturally produced CL derivative by the sulfotransferase family 2B1 (SULT2B1), which is widely present in human plasma. CS is involved in cell membrane stabilization, blood clotting, keratinocyte differentiation, and TCR nanocluster deformation. This study shows that treatment of T cells with CS resulted in the decreased surface expression of some surface T-cell proteins and reduced IL-2 release. Furthermore, T cells treated with CS significantly reduced lipid raft contents and membrane CLs. Surprisingly, using the electron microscope, we also observed that CS led to the disruption of T-cell microvilli, releasing small microvilli particles containing TCRs and other microvillar proteins. However, in vivo, T cells with CS showed aberrant migration to high endothelial venules and limited infiltrating splenic T-cell zones compared with the untreated T cells. Additionally, we observed significant alleviation of atopic dermatitis in mice injected with CS in the animal model. Based on these results, we conclude that CS is an immunosuppressive natural lipid that impairs TCR signaling by disrupting microvillar function in T cells, suggesting its usefulness as a therapeutic agent for alleviating T-cell-mediated hypersensitivity and a potential target for treating autoimmune diseases.
Collapse
Affiliation(s)
- Jeong-Su Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| | - Ik-Joo Chung
- Department of Hematology-Oncology, Immunotherapy Innovation Center, Chonnam National University Medical School, Hwasun 58128, Korea
| | - Hye-Ran Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Division of Rare and Refractory Cancer, Tumor Immunology, Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Chang-Duk Jun
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea
| |
Collapse
|
8
|
Beppler C, Eichorst J, Marchuk K, Cai E, Castellanos CA, Sriram V, Roybal KT, Krummel MF. Hyperstabilization of T cell microvilli contacts by chimeric antigen receptors. J Cell Biol 2023; 222:e202205118. [PMID: 36520493 PMCID: PMC9757849 DOI: 10.1083/jcb.202205118] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 10/25/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
T cells typically recognize their ligands using a defined cell biology-the scanning of their membrane microvilli (MV) to palpate their environment-while that same membrane scaffolds T cell receptors (TCRs) that can signal upon ligand binding. Chimeric antigen receptors (CARs) present both a therapeutic promise and a tractable means to study the interplay between receptor affinity, MV dynamics and T cell function. CARs are often built using single-chain variable fragments (scFvs) with far greater affinity than that of natural TCRs. We used high-resolution lattice lightsheet (LLS) and total internal reflection fluorescence (TIRF) imaging to visualize MV scanning in the context of variations in CAR design. This demonstrated that conventional CARs hyper-stabilized microvillar contacts relative to TCRs. Reducing receptor affinity, antigen density, and/or multiplicity of receptor binding sites normalized microvillar dynamics and synapse resolution, and effector functions improved with reduced affinity and/or antigen density, highlighting the importance of understanding the underlying cell biology when designing receptors for optimal antigen engagement.
Collapse
Affiliation(s)
- Casey Beppler
- Department of Pathology and ImmunoX, University of California, San Francisco, San Francisco, CA, USA
| | - John Eichorst
- Biological Imaging Development CoLab, University of California, San Francisco, San Francisco, CA, USA
| | - Kyle Marchuk
- Biological Imaging Development CoLab, University of California, San Francisco, San Francisco, CA, USA
| | - En Cai
- Department of Pathology and ImmunoX, University of California, San Francisco, San Francisco, CA, USA
| | - Carlos A. Castellanos
- Department of Microbiology and Immunology, Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA, USA
| | | | - Kole T. Roybal
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Helen Diller Comprehensive Cancer Center, San Francisco, CA, USA
| | - Matthew F. Krummel
- Department of Pathology and ImmunoX, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
9
|
Flowers D, Bassen D, Kapitanov GI, Marcantonio D, Burke JM, Apgar JF, Betts A, Hua F. A next generation mathematical model for the in vitro to clinical translation of T-cell engagers. J Pharmacokinet Pharmacodyn 2023; 50:215-227. [PMID: 36790614 DOI: 10.1007/s10928-023-09846-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 02/01/2023] [Indexed: 02/16/2023]
Abstract
T-cell engager (TCE) molecules activate the immune system and direct it to kill tumor cells. The key mechanism of action of TCEs is to crosslink CD3 on T cells and tumor associated antigens (TAAs) on tumor cells. The formation of this trimolecular complex (i.e. trimer) mimics the immune synapse, leading to therapeutic-dependent T-cell activation and killing of tumor cells. Computational models supporting TCE development must predict trimer formation accurately. Here, we present a next-generation two-step binding mathematical model for TCEs to describe trimer formation. Specifically, we propose to model the second binding step with trans-avidity and as a two-dimensional (2D) process where the reactants are modeled as the cell-surface density. Compared to the 3D binding model where the reactants are described in terms of concentration, the 2D model predicts less sensitivity of trimer formation to varying cell densities, which better matches changes in EC50 from in vitro cytotoxicity assay data with varying E:T ratios. In addition, when translating in vitro cytotoxicity data to predict in vivo active clinical dose for blinatumomab, the choice of model leads to a notable difference in dose prediction. The dose predicted by the 2D model aligns better with the approved clinical dose and the prediction is robust under variations in the in vitro to in vivo translation assumptions. In conclusion, the 2D model with trans-avidity to describe trimer formation is an improved approach for TCEs and is likely to produce more accurate predictions to support TCE development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Fei Hua
- Applied BioMath, Concord, MA, USA.
| |
Collapse
|
10
|
This S, Rogers D, Mallet Gauthier È, Mandl JN, Melichar HJ. What's self got to do with it: Sources of heterogeneity among naive T cells. Semin Immunol 2023; 65:101702. [PMID: 36463711 DOI: 10.1016/j.smim.2022.101702] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 12/04/2022]
Abstract
There is a long-standing assumption that naive CD4+ and CD8+ T cells are largely homogeneous populations despite the extraordinary diversity of their T cell receptors (TCR). The self-immunopeptidome plays a key role in the selection of the naive T cell repertoire in the thymus, and self-peptides are also an important driver of differences between individual naive T cells with regard to their subsequent functional contributions to an immune response. Accumulating evidence suggests that as early as the β-selection stage of T cell development, when only one of the recombined chains of the mature TCR is expressed, signaling thresholds may be established for positive selection of immature thymocytes. Stochastic encounters subsequently made with self-ligands during positive selection in the thymus imprint functional biases that a T cell will carry with it throughout its lifetime, although ongoing interactions with self in the periphery ensure a level of plasticity in the gene expression wiring of naive T cells. Identifying the sources of heterogeneity in the naive T cell population and which functional attributes of T cells can be modulated through post-thymic interventions versus those that are fixed during T cell development, could enable us to better select or generate T cells with particular traits to improve the efficacy of T cell therapies.
Collapse
Affiliation(s)
- Sébastien This
- Department of Microbiology, Infectious Disease, and Immunology, Université de Montréal, Montreal, Canada; Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montreal, Canada
| | - Dakota Rogers
- Department of Physiology and McGill Research Centre on Complex Traits, McGill University, Montreal, Canada
| | - Ève Mallet Gauthier
- Department of Microbiology, Infectious Disease, and Immunology, Université de Montréal, Montreal, Canada; Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montreal, Canada
| | - Judith N Mandl
- Department of Physiology and McGill Research Centre on Complex Traits, McGill University, Montreal, Canada.
| | - Heather J Melichar
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montreal, Canada; Department of Medicine, Université de Montréal, Montreal, Canada.
| |
Collapse
|
11
|
Britain DM, Town JP, Weiner OD. Progressive enhancement of kinetic proofreading in T cell antigen discrimination from receptor activation to DAG generation. eLife 2022; 11:e75263. [PMID: 36125261 PMCID: PMC9536835 DOI: 10.7554/elife.75263] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 09/18/2022] [Indexed: 11/16/2022] Open
Abstract
T cells use kinetic proofreading to discriminate antigens by converting small changes in antigen-binding lifetime into large differences in cell activation, but where in the signaling cascade this computation is performed is unknown. Previously, we developed a light-gated immune receptor to probe the role of ligand kinetics in T cell antigen signaling. We found significant kinetic proofreading at the level of the signaling lipid diacylglycerol (DAG) but lacked the ability to determine where the multiple signaling steps required for kinetic discrimination originate in the upstream signaling cascade (Tiseher and Weiner, 2019). Here, we uncover where kinetic proofreading is executed by adapting our optogenetic system for robust activation of early signaling events. We find the strength of kinetic proofreading progressively increases from Zap70 recruitment to LAT clustering to downstream DAG generation. Leveraging the ability of our system to rapidly disengage ligand binding, we also measure slower reset rates for downstream signaling events. These data suggest a distributed kinetic proofreading mechanism, with proofreading steps both at the receptor and at slower resetting downstream signaling complexes that could help balance antigen sensitivity and discrimination.
Collapse
Affiliation(s)
- Derek M Britain
- Cardiovascular Research Institute and Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Jason P Town
- Cardiovascular Research Institute and Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| | - Orion David Weiner
- Cardiovascular Research Institute and Department of Biochemistry and Biophysics, University of California, San FranciscoSan FranciscoUnited States
| |
Collapse
|
12
|
Samassa F, Mallone R. Self-antigens, benign autoimmunity and type 1 diabetes: a beta-cell and T-cell perspective. Curr Opin Endocrinol Diabetes Obes 2022; 29:370-378. [PMID: 35777965 DOI: 10.1097/med.0000000000000735] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW Recent work using immunopeptidomics and deconvolution of the antigenic reactivity of islet-infiltrating CD8+ T cells has expanded our knowledge about the autoimmune target epitopes of type 1 diabetes. The stem-like properties of autoimmune CD8+ T cells have also been described. We here propose a possible link between these findings. RECENT FINDINGS Weak major histocompatibility complex (MHC)-binding epitopes list among the major targets of human islet-infiltrating CD8+ T cells, likely resulting in low peptide-MHC presentation that delivers weak T-cell receptor (TCR) signals, especially in the face of low-affinity autoimmune TCRs. These weak TCR signals may favor the maintenance of the partially differentiated stem-like phenotype recently described for islet-reactive CD8+ T cells in the blood and pancreatic lymph nodes. These weak TCR signals may also be physiological, reflecting the need for self-peptide-MHC contacts to maintain homeostatic T-cell survival and proliferation. These features may underlie the universal state of benign autoimmunity that we recently described, which is characterized by islet-reactive, naïve-like CD8+ T cells circulating in all individuals. SUMMARY These observations provide novel challenges and opportunities to develop circulating T-cell biomarkers for autoimmune staging. Therapeutic halting of islet autoimmunity may require targeting of stem-like T cells to blunt their self-regeneration.
Collapse
Affiliation(s)
| | - Roberto Mallone
- Institut Cochin, Université Paris Cité, CNRS, INSERM
- Assistance Publique Hôpitaux de Paris, Service de Diabétologie et Immunologie Clinique, Cochin Hospital, Paris, France
| |
Collapse
|
13
|
Harris MJ, Chen H, Cai T, Yi Y, Deng Q, Yao Y, Lan T, Guo Y, Xu X, Wen X, McGee JE, Tatang D, Brock J, Shi F, Zhou L. Generation of Allogeneic CAR T Cells through Specific Degradation of the T Cell Antigen Receptor by E3 Ubiquitin Ligase Fusion Proteins. ACS Synth Biol 2022; 11:2029-2035. [PMID: 35549091 DOI: 10.1021/acssynbio.1c00397] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Receptor downregulation is instrumental for many therapeutic interventions. Receptor knockout through gene-editing technologies is efficient but can introduce off-target mutations and chromothripsis. Regulation of gene expression at the protein level is a promising alternative. Here, we present results showing the targeted T cell antigen receptor (TCR) degradation using chimeric E3 fusion proteins that we call Receptor Targeting Chimeras (ReceptorTAC). We show that TCR degradation is dependent on enzymatically active, membrane-anchored E3 ligase variants. TCR specificity was achieved by direct fusion of an E3 domain to the CD3ζ transmembrane sequence. Jurkat and primary T cells stably expressing the ReceptorTAC constructs showed significantly reduced responses to TCR stimulation. We also used our ReceptorTAC technology to generate TCR-deficient, claudin18.2-specific CAR T cells, where the activity of the CAR was unaffected by the expression of the ReceptorTAC. These data indicate that our ReceptorTAC molecule can be used to generate allogeneic CAR T cells.
Collapse
Affiliation(s)
- Michael J. Harris
- Boan Boston LLC, 19 Presidential Way, Suite 304, Woburn, Massachusetts 01801, United States
| | - Hao Chen
- Nanjing Boan Biotechnology Co. Ltd., 28 Gaoxin Rd., Pokou District, Nanjing, Jiangsu 210061, China
| | - Tianyu Cai
- Boan Boston LLC, 19 Presidential Way, Suite 304, Woburn, Massachusetts 01801, United States
| | - Yuting Yi
- Nanjing Boan Biotechnology Co. Ltd., 28 Gaoxin Rd., Pokou District, Nanjing, Jiangsu 210061, China
| | - Qiaowen Deng
- Nanjing Boan Biotechnology Co. Ltd., 28 Gaoxin Rd., Pokou District, Nanjing, Jiangsu 210061, China
| | - Yi Yao
- Nanjing Boan Biotechnology Co. Ltd., 28 Gaoxin Rd., Pokou District, Nanjing, Jiangsu 210061, China
| | - Tianle Lan
- Nanjing Boan Biotechnology Co. Ltd., 28 Gaoxin Rd., Pokou District, Nanjing, Jiangsu 210061, China
| | - Yanfeng Guo
- Nanjing Boan Biotechnology Co. Ltd., 28 Gaoxin Rd., Pokou District, Nanjing, Jiangsu 210061, China
| | - Xiufang Xu
- Nanjing Boan Biotechnology Co. Ltd., 28 Gaoxin Rd., Pokou District, Nanjing, Jiangsu 210061, China
| | - Xian Wen
- Nanjing Boan Biotechnology Co. Ltd., 28 Gaoxin Rd., Pokou District, Nanjing, Jiangsu 210061, China
| | - Joshua E. McGee
- Boan Boston LLC, 19 Presidential Way, Suite 304, Woburn, Massachusetts 01801, United States
| | - Daniella Tatang
- Boan Boston LLC, 19 Presidential Way, Suite 304, Woburn, Massachusetts 01801, United States
| | - James Brock
- Boan Boston LLC, 19 Presidential Way, Suite 304, Woburn, Massachusetts 01801, United States
| | - Feng Shi
- Boan Boston LLC, 19 Presidential Way, Suite 304, Woburn, Massachusetts 01801, United States
| | - Li Zhou
- Boan Boston LLC, 19 Presidential Way, Suite 304, Woburn, Massachusetts 01801, United States
| |
Collapse
|
14
|
Zhang Y, Zhu G, Li K, Li F, Huang L, Duan M, Zhou F. HLAB: learning the BiLSTM features from the ProtBert-encoded proteins for the class I HLA-peptide binding prediction. Brief Bioinform 2022; 23:6581432. [PMID: 35514183 PMCID: PMC9487590 DOI: 10.1093/bib/bbac173] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/29/2022] [Accepted: 04/18/2022] [Indexed: 12/11/2022] Open
Abstract
Human Leukocyte Antigen (HLA) is a type of molecule residing on the surfaces of most human cells and exerts an essential role in the immune system responding to the invasive items. The T cell antigen receptors may recognize the HLA-peptide complexes on the surfaces of cancer cells and destroy these cancer cells through toxic T lymphocytes. The computational determination of HLA-binding peptides will facilitate the rapid development of cancer immunotherapies. This study hypothesized that the natural language processing-encoded peptide features may be further enriched by another deep neural network. The hypothesis was tested with the Bi-directional Long Short-Term Memory-extracted features from the pretrained Protein Bidirectional Encoder Representations from Transformers-encoded features of the class I HLA (HLA-I)-binding peptides. The experimental data showed that our proposed HLAB feature engineering algorithm outperformed the existing ones in detecting the HLA-I-binding peptides. The extensive evaluation data show that the proposed HLAB algorithm outperforms all the seven existing studies on predicting the peptides binding to the HLA-A*01:01 allele in AUC and achieves the best average AUC values on the six out of the seven k-mers (k=8,9,...,14, respectively represent the prediction task of a polypeptide consisting of k amino acids) except for the 9-mer prediction tasks. The source code and the fine-tuned feature extraction models are available at http://www.healthinformaticslab.org/supp/resources.php.
Collapse
Affiliation(s)
- Yaqi Zhang
- College of Computer Science and Technology, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Gancheng Zhu
- College of Computer Science and Technology, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Kewei Li
- College of Computer Science and Technology, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Fei Li
- College of Computer Science and Technology, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Lan Huang
- College of Computer Science and Technology, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Meiyu Duan
- College of Computer Science and Technology, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Fengfeng Zhou
- College of Computer Science and Technology, and Key Laboratory of Symbolic Computation and Knowledge Engineering of Ministry of Education, Jilin University, Changchun, Jilin 130012, P.R. China
| |
Collapse
|
15
|
Morita M, Mizui M, Masuyama S, Tsokos GC, Isaka Y. Reduction of Cell Surface T-Cell Receptor by Non-Mitogenic CD3 Antibody to Mitigate Murine Lupus. Front Immunol 2022; 13:855812. [PMID: 35419004 PMCID: PMC8995471 DOI: 10.3389/fimmu.2022.855812] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 02/28/2022] [Indexed: 11/30/2022] Open
Abstract
T-cells are critically involved in the pathogenesis of systemic lupus erythematosus. Although treatment with the anti-CD3 antibody has been reported to be effective in several autoimmune disease animal models including lupus, the immunosuppressive mechanisms remain obscure because of its pleiotropic in vivo kinetics. In this study, a conventional anti-CD3 (2C11C) and a non-mitogenic anti-CD3 with a manipulated Fc region (2C11S) were compared to elucidate the underlying mechanism of action. The efficacy and safety of 2C11S in vivo were demonstrated by sustained TCR reduction for a longer period as compared to 2C11C and no induction of cytokine release or T-cell depletion. Anti-CD3s were administered to NZB/W F1 (BWF1) mice at different time points for individual periods. The short-term treatment with 2C11S in the early phase of lupus suppressed the autoantibody associated with the reduction of germinal center B-cells. Treatment in the late phase attenuated lupus nephritis without affecting autoantibodies or differentiation of effector T-cells. The effect of reduced TCR in the development of autoimmunity was examined by CD3ζ heterozygous-deficient mice, in which T-cells had reduced TCR intensity but showed normal TCR signaling response. Autoantibody and lupus nephritis were attenuated significantly in CD3ζ heterozygous-deficient lupus-prone mice. Collectively, the reduction of surface TCR by non-mitogenic anti-CD3 could sufficiently suppress the development of lupus.
Collapse
Affiliation(s)
- Masashi Morita
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Masayuki Mizui
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Satoshi Masuyama
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - George C Tsokos
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
16
|
Ahmad Z, Somanath PR. AKT Isoforms in the Immune Response in Cancer. Curr Top Microbiol Immunol 2022; 436:349-366. [PMID: 36243852 DOI: 10.1007/978-3-031-06566-8_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
AKT is a protein kinase that exists in three isoforms: AKT1, AKT2, and AKT3. Though similar in structure, these isoforms display different effects. AKT is activated downstream of PI3K, and together, this signaling pathway helps regulate cellular processes including cell growth, proliferation, metabolism, survival, and apoptosis. Disruption in these pathways has been associated with disorders including cardiovascular diseases, developmental disorders, inflammatory responses, autoimmune diseases, neurologic disorders, type 2 diabetes, and several cancers. In cancer, deregulation in the PI3K/AKT pathway can be manifested as tumorigenesis, pathological angiogenesis, and metastasis. Increased activity has been correlated with tumor progression and resistance to cancer treatments. Recent studies have suggested that inhibition of the PI3K/AKT pathway plays a significant role in the development, expansion, and proliferation of cells of the immune system. Additionally, AKT has been found to play an important role in differentiating regulatory T cells, activating B cells, and augmenting tumor immunosurveillance. This emphasizes AKT as a potential target for inhibition in cancer therapy. This chapter reviews AKT structure and regulation, its different isoforms, its role in immune cells, and its modulation in oncotherapy.
Collapse
Affiliation(s)
- Zayd Ahmad
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA
| | - Payaningal R Somanath
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA, 30912, USA.
- Georgia Cancer Center, Vascular Biology Center and Department of Medicine, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
17
|
Abstract
Conventional CD4+ and CD8+ T lymphocytes comprise a mixture of naive and memory cells. Generation and survival of these T-cell subsets is under strict homeostatic control and reflects contact with self-major histocompatibility complex (MHC) and certain cytokines. Naive T cells arise in the thymus via T-cell receptor (TCR)-dependent positive selection to self-peptide/MHC complexes and are then maintained in the periphery through self-MHC interaction plus stimulation via interleukin-7 (IL-7). By contrast, memory T cells are largely MHC-independent for their survival but depend strongly on stimulation via cytokines. Whereas typical memory T cells are generated in response to foreign antigens, some arise spontaneously through contact of naive precursors with self-MHC ligands; we refer to these cells as memory-phenotype (MP) T cells. In this review, we discuss the generation and homeostasis of naive T cells and these two types of memory T cells, focusing on their relative interaction with MHC ligands and cytokines.
Collapse
Affiliation(s)
- Takeshi Kawabe
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Jaeu Yi
- Division of Rheumatology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Jonathan Sprent
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
- St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales 2010, Australia
| |
Collapse
|
18
|
Efficient killing of tumor cells by CAR-T cells demands engagement of a larger number of CARs as opposed to TCRs. J Biol Chem 2021; 297:101033. [PMID: 34371020 PMCID: PMC8452787 DOI: 10.1016/j.jbc.2021.101033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 01/21/2023] Open
Abstract
Although CAR T cells are widely used to treat cancer, efficiency of CAR-T cell cytolytic responses has not been carefully examined. We engineered CAR specific for HMW-MAA (high molecular weight melanoma-associated antigen) and evaluated potency of CD8+ CAR-T cells to release cytolytic granules and to kill tissue-derived melanoma cells, which express different levels of HMW-MAA. CAR T cells efficiently killed melanoma cells expressing high level of HMW-MAA, but not melanoma cells with lower levels of HMW-MAA. The same melanoma cells presenting significantly lower level of stimulatory peptide-MHC ligand were readily lysed by T cells transduced with genes encoding α,β-TCR specific for the peptide-MHC ligand. The data suggest that higher level of targeted molecules is required to engage a larger number of CARs than TCRs to induce efficient cytolytic granule release and destruction of melanoma cells. Understanding the difference in molecular mechanisms controlling activation thresholds of CAR- versus TCR-mediated responses will contribute to improving efficiency of CAR T cells required to eliminate solid tumors presenting low levels of targeted molecules.
Collapse
|
19
|
Bezverbnaya K, Moogk D, Cummings D, Baker CL, Aarts C, Denisova G, Sun M, McNicol JD, Turner RC, Rullo AF, Foley SR, Bramson JL. Development of a B-cell maturation antigen-specific T-cell antigen coupler receptor for multiple myeloma. Cytotherapy 2021; 23:820-832. [PMID: 34217618 DOI: 10.1016/j.jcyt.2021.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/13/2021] [Accepted: 05/14/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND AIMS T cells engineered with synthetic receptors have delivered powerful therapeutic results for patients with relapsed/refractory hematologic malignancies. The authors have recently described the T-cell antigen coupler (TAC) receptor, which co-opts the endogenous T-cell receptor (TCR) and activates engineered T cells in an HLA-independent manner. Here the authors describe the evolution of a next-generation TAC receptor with a focus on developing a TAC-engineered T cell for multiple myeloma. METHODS To optimize the TAC scaffold, the authors employed a bona fide antigen-binding domain derived from the B-cell maturation antigen-specific monoclonal antibody C11D5.3, which has been used successfully in the clinic. The authors first tested humanized versions of the UCHT1 domain, which is used by the TAC to co-opt the TCR. The authors further discovered that the signal peptide affected surface expression of the TAC receptor. Higher density of the TAC receptor enhanced target binding in vitro, which translated into higher levels of Lck at the immunological synapse and stronger proliferation when only receptor-ligand interactions were present. RESULTS The authors observed that the humanized UCHT1 improved surface expression and in vivo efficacy. Using TAC T cells derived from both healthy donors and multiple myeloma patients, the authors determined that despite the influence of receptor density on early activation events and effector function, receptor density did not impact late effector functions in vitro, nor did the receptor density affect in vivo efficacy. CONCLUSIONS The modifications to the TAC scaffold described herein represent an important step in the evolution of this technology, which tolerates a range of expression levels without impacting therapeutic efficacy.
Collapse
Affiliation(s)
- Ksenia Bezverbnaya
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Duane Moogk
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Derek Cummings
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Christopher L Baker
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Craig Aarts
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Galina Denisova
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Michael Sun
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Jamie D McNicol
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Rebecca C Turner
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - Anthony F Rullo
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada
| | - S Ronan Foley
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada; Juravinski Hospital and Cancer Centre, Hamilton, Canada
| | - Jonathan L Bramson
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, Canada.
| |
Collapse
|
20
|
Trefzer A, Kadam P, Wang SH, Pennavaria S, Lober B, Akçabozan B, Kranich J, Brocker T, Nakano N, Irmler M, Beckers J, Straub T, Obst R. Dynamic adoption of anergy by antigen-exhausted CD4 + T cells. Cell Rep 2021; 34:108748. [PMID: 33567282 DOI: 10.1016/j.celrep.2021.108748] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 09/21/2020] [Accepted: 01/19/2021] [Indexed: 12/11/2022] Open
Abstract
Exhausted immune responses to chronic diseases represent a major challenge to global health. We study CD4+ T cells in a mouse model with regulatable antigen presentation. When the cells are driven through the effector phase and are then exposed to different levels of persistent antigen, they lose their T helper 1 (Th1) functions, upregulate exhaustion markers, resemble naturally anergic cells, and modulate their MAPK, mTORC1, and Ca2+/calcineurin signaling pathways with increasing dose and time. They also become unable to help B cells and, at the highest dose, undergo apoptosis. Transcriptomic analyses show the dynamic adjustment of gene expression and the accumulation of T cell receptor (TCR) signals over a period of weeks. Upon antigen removal, the cells recover their functionality while losing exhaustion and anergy markers. Our data suggest an adjustable response of CD4+ T cells to different levels of persisting antigen and contribute to a better understanding of chronic disease.
Collapse
Affiliation(s)
- Anne Trefzer
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Pallavi Kadam
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Shu-Hung Wang
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Stefanie Pennavaria
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Benedikt Lober
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Batuhan Akçabozan
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Jan Kranich
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Thomas Brocker
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Naoko Nakano
- Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba 278-0022, Japan
| | - Martin Irmler
- Institute of Experimental Genetics, Helmholtz Zentrum München GmbH, 85764 Neuherberg, Germany
| | - Johannes Beckers
- Institute of Experimental Genetics, Helmholtz Zentrum München GmbH, 85764 Neuherberg, Germany; Chair of Experimental Genetics, Technische Universität München, 85354 Freising, Germany; German Center for Diabetes Research (DZD e. V.), Neuherberg, Germany
| | - Tobias Straub
- Bioinformatics Core Facility, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany
| | - Reinhard Obst
- Institute for Immunology, Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany.
| |
Collapse
|
21
|
Siokis A, Robert PA, Meyer-Hermann M. Agent-Based Modeling of T Cell Receptor Cooperativity. Int J Mol Sci 2020; 21:ijms21186473. [PMID: 32899840 PMCID: PMC7555007 DOI: 10.3390/ijms21186473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 11/25/2022] Open
Abstract
Immunological synapse (IS) formation is a key event during antigen recognition by T cells. Recent experimental evidence suggests that the affinity between T cell receptors (TCRs) and antigen is actively modulated during the early steps of TCR signaling. In this work, we used an agent-based model to study possible mechanisms for affinity modulation during IS formation. We show that, without any specific active mechanism, the observed affinity between receptors and ligands evolves over time and depends on the density of ligands of the antigen peptide presented by major histocompatibility complexes (pMHC) and TCR molecules. A comparison between the presence or absence of TCR–pMHC centrally directed flow due to F-actin coupling suggests that centripetal transport is a potential mechanism for affinity modulation. The model further suggests that the time point of affinity measurement during immune synapse formation is critical. Finally, a mathematical model of F-actin foci formation incorporated in the agent-based model shows that TCR affinity can potentially be actively modulated by positive/negative feedback of the F-actin foci on the TCR-pMHC association rate kon.
Collapse
Affiliation(s)
- Anastasios Siokis
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, 38106 Braunschweig, Germany; (A.S.); (P.A.R.)
| | - Philippe A. Robert
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, 38106 Braunschweig, Germany; (A.S.); (P.A.R.)
| | - Michael Meyer-Hermann
- Department of Systems Immunology and Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, 38106 Braunschweig, Germany; (A.S.); (P.A.R.)
- Institute of Biochemistry, Biotechnology and Bioinformatics, Technische Universität Braunschweig, 38106 Braunschweig, Germany
- Correspondence: ; Tel.: +49-531-391-55210
| |
Collapse
|
22
|
Sng XYX, Li J, Zareie P, Assmus LM, Lee JKC, Jones CM, Turner SJ, Daley SR, Quinn KM, La Gruta NL. The Impact of MHC Class I Dose on Development and Maintenance of the Polyclonal Naive CD8+ T Cell Repertoire. THE JOURNAL OF IMMUNOLOGY 2020; 204:3108-3116. [DOI: 10.4049/jimmunol.2000081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/02/2020] [Indexed: 12/14/2022]
|
23
|
Porsche CE, Delproposto JB, Patrick E, Zamarron BF, Lumeng CN. Adipose tissue dendritic cell signals are required to maintain T cell homeostasis and obesity-induced expansion. Mol Cell Endocrinol 2020; 505:110740. [PMID: 31987897 PMCID: PMC7197735 DOI: 10.1016/j.mce.2020.110740] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/22/2020] [Accepted: 01/22/2020] [Indexed: 02/06/2023]
Abstract
Adipose tissue derived chronic inflammation is a critical component of obesity induced type II diabetes. Major histocompatibility complex II (MHCII) mediated T cell activation within adipose tissue is one mechanism that contributes to this phenotype. However, the contribution of dendritic cells as professional antigen presenting cells in adipose issue has not previously been explored. Using ItgaxCre x MHCIIfl/fl (M11cKO) mice we observed adipose tissue specific changes in adipose tissue leukocytes. While there was a complete knockout of MHCII in dendritic cells, MHCII was also absent on the majority of macrophages. This resulted in reduction of TCR expression in CD4+ T cells in obese adipose tissue, and an increase in CD8+ and CD4+ CD8+ double positive T cells with decreased CD4+ T cells independent of diet type. Increased CD8+ cells were not observed in the spleen, suggesting adipose tissue T cell regulation is tissue specific. In vitro studies demonstrated more potent antigen presentation function in adipose tissue dendritic cells compared to macrophages. Obese M11cKO mice had decreased CD11c+ adipose tissue macrophages. Despite the changes of immune cellularity in adipose tissue, M11cKO largely did not change inflammatory gene expression in adipose tissue and did not demonstrate differences in glucose and insulin intolerance. Overall MHCII expression on CD11c+ cells is important for maintaining CD4+ and CD8+ adipose tissue T cells, but these cellular changes fail to alter inflammatory output and systemic metabolism.
Collapse
Affiliation(s)
- Cara E Porsche
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jennifer B Delproposto
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Elise Patrick
- College of Literature Sciences and Arts, University of Michigan, Ann Arbor, USA
| | - Brian F Zamarron
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Carey N Lumeng
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Pediatrics and Communicable Diseases, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
24
|
He Q, Jiang X, Zhou X, Weng J. Targeting cancers through TCR-peptide/MHC interactions. J Hematol Oncol 2019; 12:139. [PMID: 31852498 PMCID: PMC6921533 DOI: 10.1186/s13045-019-0812-8] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 10/27/2019] [Indexed: 02/07/2023] Open
Abstract
Adoptive T cell therapy has achieved dramatic success in a clinic, and the Food and Drug Administration approved two chimeric antigen receptor-engineered T cell (CAR-T) therapies that target hematological cancers in 2018. A significant issue faced by CAR-T therapies is the lack of tumor-specific biomarkers on the surfaces of solid tumor cells, which hampers the application of CAR-T therapies to solid tumors. Intracellular tumor-related antigens can be presented as peptides in the major histocompatibility complex (MHC) on the cell surface, which interact with the T cell receptors (TCR) on antigen-specific T cells to stimulate an anti-tumor response. Multiple immunotherapy strategies have been developed to eradicate tumor cells through targeting the TCR-peptide/MHC interactions. Here, we summarize the current status of TCR-based immunotherapy strategies, with particular focus on the TCR structure, activated signaling pathways, the effects and toxicity associated with TCR-based therapies in clinical trials, preclinical studies examining immune-mobilizing monoclonal TCRs against cancer (ImmTACs), and TCR-fusion molecules. We propose several TCR-based therapeutic strategies to achieve optimal clinical responses without the induction of autoimmune diseases.
Collapse
Affiliation(s)
- Qinghua He
- Department of Center Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, 621 Gangwan Rd, Huangpu Qu, Guangzhou, 510700, China
| | - Xianhan Jiang
- Department of General Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China
| | - Xinke Zhou
- Department of Center Laboratory, The Fifth Affiliated Hospital of Guangzhou Medical University, 621 Gangwan Rd, Huangpu Qu, Guangzhou, 510700, China. .,Department of General Surgery, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510700, China.
| | - Jinsheng Weng
- Department of Lymphoma and Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1414 Holcombe Boulevard, Houston, TX, 77030, USA.
| |
Collapse
|
25
|
Thomas S, Mohammed F, Reijmers RM, Woolston A, Stauss T, Kennedy A, Stirling D, Holler A, Green L, Jones D, Matthews KK, Price DA, Chain BM, Heemskerk MHM, Morris EC, Willcox BE, Stauss HJ. Framework engineering to produce dominant T cell receptors with enhanced antigen-specific function. Nat Commun 2019; 10:4451. [PMID: 31575864 PMCID: PMC6773850 DOI: 10.1038/s41467-019-12441-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 08/26/2019] [Indexed: 12/28/2022] Open
Abstract
TCR-gene-transfer is an efficient strategy to produce therapeutic T cells of defined antigen specificity. However, there are substantial variations in the cell surface expression levels of human TCRs, which can impair the function of engineered T cells. Here we demonstrate that substitutions of 3 amino acid residues in the framework of the TCR variable domains consistently increase the expression of human TCRs on the surface of engineered T cells.The modified TCRs mediate enhanced T cell proliferation, cytokine production and cytotoxicity, while reducing the peptide concentration required for triggering effector function up to 3000-fold. Adoptive transfer experiments in mice show that modified TCRs control tumor growth more efficiently than wild-type TCRs. Our data indicate that simple variable domain modifications at a distance from the antigen-binding loops lead to increased TCR expression and improved effector function. This finding provides a generic platform to optimize the efficacy of TCR gene therapy in humans.
Collapse
MESH Headings
- Animals
- Antigens/immunology
- Antigens, CD/metabolism
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Cell Engineering
- Cell Line, Tumor
- Cell Proliferation
- Cytokines/metabolism
- Gene Expression
- Genes, T-Cell Receptor/genetics
- Genes, T-Cell Receptor/immunology
- Genetic Therapy
- Humans
- Lectins, C-Type/metabolism
- Lymphocyte Activation
- Male
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Models, Molecular
- Protein Domains
- Protein Engineering
- Receptors, Antigen, T-Cell/chemistry
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Sharyn Thomas
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, London, NW3 2PF, UK
| | - Fiyaz Mohammed
- Cancer Immunology and Immunotherapy Centre, Institute for Immunology and Immunotherapy, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Rogier M Reijmers
- Department of Hematology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Annemarie Woolston
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, London, NW3 2PF, UK
| | - Theresa Stauss
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, London, NW3 2PF, UK
| | - Alan Kennedy
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, London, NW3 2PF, UK
| | - David Stirling
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, London, NW3 2PF, UK
| | - Angelika Holler
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, London, NW3 2PF, UK
| | - Louisa Green
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, London, NW3 2PF, UK
| | - David Jones
- Department of Computer Science, University College London, London, WC1E 6BT, UK
| | - Katherine K Matthews
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, CF10 3AT, UK
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, CF10 3AT, UK
| | - Benjamin M Chain
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, London, NW3 2PF, UK
| | - Mirjam H M Heemskerk
- Department of Hematology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Emma C Morris
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, London, NW3 2PF, UK
| | - Benjamin E Willcox
- Cancer Immunology and Immunotherapy Centre, Institute for Immunology and Immunotherapy, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Hans J Stauss
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, London, NW3 2PF, UK.
| |
Collapse
|
26
|
Impact of epitope density on CD8+ T cell development and function. Mol Immunol 2019; 113:120-125. [DOI: 10.1016/j.molimm.2019.03.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/17/2019] [Accepted: 03/21/2019] [Indexed: 11/23/2022]
|
27
|
Abstract
Generating and maintaining a diverse repertoire of naive T cells is essential for protection against pathogens, and developing a mechanistic and quantitative description of the processes involved lies at the heart of our understanding of vertebrate immunity. Here, we review the biology of naive T cells from birth to maturity and outline how the integration of mathematical models and experiments has helped us to develop a full picture of their life histories.
Collapse
Affiliation(s)
- Benedict Seddon
- Institute of Immunity and Transplantation, Division of Infection and Immunity, UCL, Royal Free Hospital, London, UK
| | - Andrew J Yates
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, New York
| |
Collapse
|
28
|
Mesangial Cells Exhibit Features of Antigen-Presenting Cells and Activate CD4+ T Cell Responses. J Immunol Res 2019; 2019:2121849. [PMID: 31317046 PMCID: PMC6604415 DOI: 10.1155/2019/2121849] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 04/01/2019] [Accepted: 05/14/2019] [Indexed: 01/18/2023] Open
Abstract
Background Mesangial cells play a prominent role in the development of inflammatory diseases and autoimmune disorders of the kidney. Mesangial cells perform the essential functions of helping to ensure that the glomerular structure is stable and regulating capillary flow, and activated mesangial cells acquire proinflammatory activities. We investigated whether activated mesangial cells display immune properties and control the development of T cell immunity. Methods Flow cytometry analysis was used to study the expression of antigen-presenting cell surface markers and costimulatory molecules in mesangial cells. CD4+ T cell activation induced by mesangial cells was detected in terms of T cell proliferation and cytokine production. Results IFN-γ-treated mesangial cells express membrane proteins involved in antigen presentation and T cell activation, including MHC-II, ICAM-1, CD40, and CD80. This finding suggests that activated mesangial cells can take up and present antigenic peptides to initiate CD4+ T cell responses and thus act as nonprofessional antigen-presenting cells. Polarization of naïve CD4+ T cells (Th0 cells) towards the Th1 phenotype was induced by coculture with activated mesangial cells, and the resulting Th1 cells showed increased mRNA and protein expression of inflammation-associated genes. Conclusion Mesangial cells can present antigen and modulate CD4+ T lymphocyte proliferation and differentiation. Interactions between mesangial cells and T cells are essential for sustaining the inflammatory response in a variety of glomerulonephritides. Therefore, mesangial cells might participate in immune function in the kidney.
Collapse
|
29
|
Gupta PK, McIntosh CM, Chong AS, Alegre ML. The pursuit of transplantation tolerance: new mechanistic insights. Cell Mol Immunol 2019; 16:324-333. [PMID: 30760917 DOI: 10.1038/s41423-019-0203-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 01/17/2019] [Indexed: 12/11/2022] Open
Abstract
Donor-specific transplantation tolerance that enables weaning from immunosuppressive drugs but retains immune competence to non-graft antigens has been a lasting pursuit since the discovery of neonatal tolerance. More recently, efforts have been devoted not only to understanding how transplantation tolerance can be induced but also the mechanisms necessary to maintain it as well as how inflammatory exposure challenges its durability. This review focuses on recent advances regarding key peripheral mechanisms of T cell tolerance, with the underlying hypothesis that a combination of several of these mechanisms may afford a more robust and durable tolerance and that a better understanding of these individual pathways may permit longitudinal tracking of tolerance following clinical transplantation to serve as biomarkers. This review may enable a personalized assessment of the degree of tolerance in individual patients and the opportunity to strengthen the robustness of peripheral tolerance.
Collapse
Affiliation(s)
- Pawan K Gupta
- Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA
| | | | - Anita S Chong
- Department of Surgery, The University of Chicago, Chicago, IL, 60637, USA
| | - Maria-Luisa Alegre
- Department of Medicine, The University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
30
|
Schuldt NJ, Binstadt BA. Dual TCR T Cells: Identity Crisis or Multitaskers? JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 202:637-644. [PMID: 30670579 PMCID: PMC11112972 DOI: 10.4049/jimmunol.1800904] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 08/21/2018] [Indexed: 05/25/2024]
Abstract
Dual TCR T cells are a common and natural product of TCR gene rearrangement and thymocyte development. As much as one third of the T cell population may have the capability to express two different TCR specificities on the cell surface. This discovery provoked a reconsideration of the classic model of thymic selection. Many potential roles for dual TCR T cells have since been hypothesized, including posing an autoimmune hazard, dominating alloreactive T cell responses, inducing allergy, and expanding the TCR repertoire to improve protective immunity. Yet, since the initial wave of publications following the discovery of dual TCR T cells, research in the area has slowed. In this study, we aim to provide a brief but comprehensive history of dual TCR T cell research, re-evaluate past observations in the context of current knowledge of the immune system, and identify key issues for future study.
Collapse
Affiliation(s)
- Nathaniel J Schuldt
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454; and Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Bryce A Binstadt
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454; and Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
31
|
Gauthier SD, Moutuou MM, Daudelin F, Leboeuf D, Guimond M. IL-7 Is the Limiting Homeostatic Factor that Constrains Homeostatic Proliferation of CD8 + T Cells after Allogeneic Stem Cell Transplantation and Graft-versus-Host Disease. Biol Blood Marrow Transplant 2018; 25:648-655. [PMID: 30576835 DOI: 10.1016/j.bbmt.2018.12.066] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 12/11/2018] [Indexed: 11/29/2022]
Abstract
Immune reconstitution after allogeneic hematopoietic stem cell transplantation relies primarily on homeostatic proliferation (HP) of mature T lymphocytes, but this process is typically impaired during graft-versus-host disease (GVHD). We previously showed that low IL-7 levels combined with lack of dendritic cell (DC) regeneration constrain CD4+ T cell HP during GVHD. However, it is not clear whether these alterations to the peripheral CD4+ T cell niche also contribute to impair CD8+ T cell regeneration during GVHD. We found that IL-7 therapy was sufficient for restoring CD8+ T cell HP in GVHD hosts while forcing DC regeneration with Flt3-L had only a modest effect on CD8+ T cell HP in IL-7 treated mice. Using bone marrow chimeras, we showed that HP of naïve CD8+ T cells is primarily regulated by MHC class I on radio-resistant stromal cells, yet optimal recovery of CD8+ T cell counts still requires expression of MHC class I on both radio-resistant and radio-sensitive hematopoietic cells. Thus, IL-7 level is the primary limiting factor that constrains naïve CD8+ T cell HP during GVHD, and accessibility of MHC class I on stromal cells explains how IL-7 therapy, as a single agent, can induce robust CD8 + T cell HP in the absence of DCs.
Collapse
Affiliation(s)
- Simon-David Gauthier
- Départment de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Moutuaata M Moutuou
- Départment de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Francis Daudelin
- Départment de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Dominique Leboeuf
- Division d'Hématologie-Oncologie, Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada; Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Martin Guimond
- Départment de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Québec, Canada; Division d'Hématologie-Oncologie, Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada.
| |
Collapse
|
32
|
Rahman A, Tiwari A, Narula J, Hickling T. Importance of Feedback and Feedforward Loops to Adaptive Immune Response Modeling. CPT Pharmacometrics Syst Pharmacol 2018; 7:621-628. [PMID: 30198637 PMCID: PMC6202469 DOI: 10.1002/psp4.12352] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 08/15/2018] [Indexed: 12/15/2022] Open
Abstract
The human adaptive immune system is a very complex network of different types of cells, cytokines, and signaling molecules. This complex network makes it difficult to understand the system level regulations. To properly explain the immune system, it is necessary to explicitly investigate the presence of different feedback and feedforward loops (FFLs) and their crosstalks. Considering that these loops increase the complexity of the system, the mathematical modeling has been proved to be an important tool to explain such complex biological systems. This review focuses on these regulatory loops and discusses their importance on systems modeling of the immune system.
Collapse
|
33
|
Augmentation of Antitumor Immunity by Human and Mouse CAR T Cells Secreting IL-18. Cell Rep 2018; 20:3025-3033. [PMID: 28954221 PMCID: PMC6002762 DOI: 10.1016/j.celrep.2017.09.002] [Citation(s) in RCA: 369] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 07/16/2017] [Accepted: 08/30/2017] [Indexed: 12/19/2022] Open
Abstract
The effects of transgenically encoded human and mouse IL-18 on T cell proliferation and its application in boosting chimeric antigen receptor (CAR) T cells are presented. Robust enhancement of proliferation of IL-18-secreting human T cells occurred in a xenograft model, and this was dependent on TCR and IL-18R signaling. IL-18 augmented IFN-γ secretion and proliferation of T cells activated by the endogenous TCR. TCR-deficient, human IL-18-expressing CD19 CAR T cells exhibited enhanced proliferation and antitumor activity in the xenograft model. Antigen-propelled activation of cytokine helper ensemble (APACHE) CAR T cells displayed inducible expression of IL-18 and enhanced antitumor immunity. In an intact mouse tumor model, CD19-IL-18 CAR T cells induced deeper B cell aplasia, significantly enhanced CAR T cell proliferation, and effectively augmented antitumor effects in mice with B16F10 melanoma. These findings point to a strategy to develop universal CAR T cells for patients with solid tumors.
Collapse
|
34
|
Borlido J, Sakuma S, Raices M, Carrette F, Tinoco R, Bradley LM, D'Angelo MA. Nuclear pore complex-mediated modulation of TCR signaling is required for naïve CD4 + T cell homeostasis. Nat Immunol 2018; 19:594-605. [PMID: 29736031 PMCID: PMC5976539 DOI: 10.1038/s41590-018-0103-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 03/28/2018] [Indexed: 11/09/2022]
Abstract
Nuclear pore complexes (NPCs) are channels connecting the nucleus with the cytoplasm. We report that loss of the tissue-specific NPC component Nup210 causes a severe deficit of naïve CD4+ T cells. Nup210-deficient CD4+ T lymphocytes develop normally but fail to survive in the periphery. The decreased survival results from both an impaired ability to transmit tonic T cell receptor (TCR) signals and increased levels of Fas, which sensitize Nup210-/- naïve CD4+ T cells to Fas-mediated cell death. Mechanistically, Nup210 regulates these processes by modulating the expression of Cav2 (encoding Caveolin-2) and Jun at the nuclear periphery. Whereas the TCR-dependent and CD4+ T cell-specific upregulation of Cav2 is critical for proximal TCR signaling, cJun expression is required for STAT3-dependent repression of Fas. Our results uncover an unexpected role for Nup210 as a cell-intrinsic regulator of TCR signaling and T cell homeostasis and expose NPCs as key players in the adaptive immune system.
Collapse
Affiliation(s)
- Joana Borlido
- Development, Aging and Regeneration Program and NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Stephen Sakuma
- Development, Aging and Regeneration Program and NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Marcela Raices
- Development, Aging and Regeneration Program and NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Florent Carrette
- Infectious and Inflammatory Disease Center and NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Roberto Tinoco
- Infectious and Inflammatory Disease Center and NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Linda M Bradley
- Infectious and Inflammatory Disease Center and NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Maximiliano A D'Angelo
- Development, Aging and Regeneration Program and NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
- Infectious and Inflammatory Disease Center and NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
35
|
An optimized single chain TCR scaffold relying on the assembly with the native CD3-complex prevents residual mispairing with endogenous TCRs in human T-cells. Oncotarget 2018; 7:21199-221. [PMID: 27028870 PMCID: PMC5008279 DOI: 10.18632/oncotarget.8385] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 03/10/2016] [Indexed: 12/29/2022] Open
Abstract
Immunotherapy of cancer envisions the adoptive transfer of T-cells genetically engineered with tumor-specific heterodimeric α/β T-cell receptors (TCRα/β). However, potential mispairing of introduced TCRα/β-chains with endogenous β/α-ones may evoke unpredictable autoimmune reactivities. A novel single chain (sc)TCR format relies on the fusion of the Vα-Linker-Vβ-fragment to the TCR Cβ-domain and coexpression of the TCR Cα-domain capable of recruiting the natural CD3-complex for full and hence, native T-cell signaling. Here, we tested whether such a gp100(280-288)- or p53(264-272) tumor antigen-specific scTCR is still prone to mispairing with TCRα. In a human Jurkat-76 T-cell line lacking endogenous TCRs, surface expression and function of a scTCR could be reconstituted by any cointroduced TCRα-chain indicating mispairing to take place on a molecular basis. In contrast, transduction into human TCRα/β-positive T-cells revealed that mispairing is largely reduced. Competition experiments in Jurkat-76 confirmed the preference of dcTCR to selfpair and to spare scTCR. This also allowed for the generation of dc/scTCR-modified cytomegalovirus/tumor antigen-bispecific T-cells to augment T-cell activation in CMV-infected tumor patients. Residual mispairing was prevented by strenghtening the Vα-Li-Vβ-fragment through the design of a novel disulfide bond between a Vα- and a linker-resident residue close to Vβ. Multimer-stainings, and cytotoxicity-, IFNγ-secretion-, and CFSE-proliferation-assays, the latter towards dendritic cells endogenously processing RNA-electroporated gp100 antigen proved the absence of hybrid scTCR/TCRα-formation without impairing avidity of scTCR/Cα in T-cells. Moreover, a fragile cytomegalovirus pp65(495-503)-specific scTCR modified this way acquired enhanced cytotoxicity. Thus, optimized scTCR/Cα inhibits residual TCR mispairing to accomplish safe adoptive immunotherapy for bulk endogenous TCRα/β-positive T-cells.
Collapse
|
36
|
Stockslager MA, Bagnall JS, Hecht VC, Hu K, Aranda-Michel E, Payer K, Kimmerling RJ, Manalis SR. Microfluidic platform for characterizing TCR-pMHC interactions. BIOMICROFLUIDICS 2017; 11:064103. [PMID: 29204244 PMCID: PMC5685811 DOI: 10.1063/1.5002116] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 10/20/2017] [Indexed: 05/03/2023]
Abstract
The physical characteristics of the T cell receptor (TCR)-peptide-major histocompatibility complex (pMHC) interaction are known to play a central role in determining T cell function in the initial stages of the adaptive immune response. State-of-the-art assays can probe the kinetics of this interaction with single-molecular-bond resolution, but this precision typically comes at the cost of low throughput, since the complexity of these measurements largely precludes "scaling up." Here, we explore the feasibility of detecting specific TCR-pMHC interactions by flowing T cells past immobilized pMHC and measuring the reduction in cell speed due to the mechanical force of the receptor-ligand interaction. To test this new fluidic measurement modality, we fabricated a microfluidic device in which pMHC-coated beads are immobilized in hydrodynamic traps along the length of a serpentine channel. As T cells flow past the immobilized beads, their change in speed is tracked via microscopy. We validated this approach using two model systems: primary CD8+ T cells from an OT-1 TCR transgenic mouse with beads conjugated with H-2Kb:SIINFEKL, and Jurkat T cells with beads conjugated with anti-CD3 and anti-CD28 antibodies.
Collapse
Affiliation(s)
- Max A Stockslager
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Josephine Shaw Bagnall
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Vivian C Hecht
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Kevin Hu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Edgar Aranda-Michel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Kristofor Payer
- Microsystems Technology Laboratories, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Robert J Kimmerling
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | |
Collapse
|
37
|
DNA-binding of the Tet-transactivator curtails antigen-induced lymphocyte activation in mice. Nat Commun 2017; 8:1028. [PMID: 29044097 PMCID: PMC5647323 DOI: 10.1038/s41467-017-01022-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 08/14/2017] [Indexed: 12/31/2022] Open
Abstract
The Tet-On/Off system for conditional transgene expression constitutes state-of-the-art technology to study gene function by facilitating inducible expression in a timed and reversible manner. Several studies documented the suitability and versatility of this system to trace lymphocyte fate and to conditionally express oncogenes or silence tumour suppressor genes in vivo. Here, we show that expression of the tetracycline/doxycycline-controlled Tet-transactivator, while tolerated well during development and in immunologically unchallenged animals, impairs the expansion of antigen-stimulated T and B cells and thereby curtails adaptive immune responses in vivo. Transactivator-mediated cytotoxicity depends on DNA binding, but can be overcome by BCL2 overexpression, suggesting that apoptosis induction upon lymphocyte activation limits cellular and humoral immune responses. Our findings suggest a possible system-intrinsic biological bias of the Tet-On/Off system in vivo that will favour the outgrowth of apoptosis resistant clones, thus possibly confounding data published using such systems. Tet-transactivators are used for direct regulation of gene expression, RNA interference and for CRISPR/Cas9-based systems. Here the authors show that DNA-bound Tet-transactivators can induce cell death in antigen-activated lymphocytes in vivo, putting into question the use of, and in vivo data generated with, these molecular tools.
Collapse
|
38
|
Wu S, Zhu W, Peng Y, Wang L, Hong Y, Huang L, Dong D, Xie J, Merchen T, Kruse E, Guo ZS, Bartlett D, Fu N, He Y. The Antitumor Effects of Vaccine-Activated CD8 + T Cells Associate with Weak TCR Signaling and Induction of Stem-Like Memory T Cells. Cancer Immunol Res 2017; 5:908-919. [PMID: 28851693 DOI: 10.1158/2326-6066.cir-17-0016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 05/02/2017] [Accepted: 08/22/2017] [Indexed: 02/04/2023]
Abstract
To understand why vaccine-activated tumor-specific T cells often fail to generate antitumor effects, we studied two α-fetoprotein-specific CD8+ T cells (Tet499 and Tet212) that had different antitumor effects. We found that Tet499 required high antigen doses for reactivation, but could survive persistent antigen stimulation and maintain their effector functions. In contrast, Tet212 had a low threshold of reactivation, but underwent exhaustion and apoptosis in the presence of persistent antigen. In vivo, Tet499 cells expanded more than Tet212 upon reencountering antigen and generated stronger antitumor effects. The different antigen responsiveness and antitumor effects of Tet212 and Tet499 cells correlated with their activation and differentiation states. Compared with Tet212, the population of Tet499 cells was less activated and contained more stem-like memory T cells (Tscm) that could undergo expansion in vivo The TCR signaling strength on Tet499 was weaker than Tet212, correlating with more severe Tet499 TCR downregulation. Weak TCR signaling may halt T-cell differentiation at the Tscm stage during immune priming and also explains why Tet499 reactivation requires a high antigen dose. Weak TCR signaling of Tet499 cells in the effector stage will also protect them from exhaustion and apoptosis when they reencounter persistent antigen in tumor lesion, which generates antitumor effects. Further investigation of TCR downregulation and manipulation of TCR signaling strength may help design cancer vaccines to elicit a mix of tumor-specific CD8+ T cells, including Tscm, capable of surviving antigen restimulation to generate antitumor effects. Cancer Immunol Res; 5(10); 908-19. ©2017 AACR.
Collapse
Affiliation(s)
- Sha Wu
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia.,Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Wei Zhu
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia.,Division of Laboratory Medicine of Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yibing Peng
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Lan Wang
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Yuan Hong
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Lei Huang
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Dayong Dong
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Junping Xie
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Todd Merchen
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia.,Department of Surgery, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Edward Kruse
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia.,Department of Surgery, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Zong Sheng Guo
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - David Bartlett
- University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania
| | - Ning Fu
- Division of Laboratory Medicine of Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Yukai He
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia. .,Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia
| |
Collapse
|
39
|
Tokarz-Deptuła B, Malinowska M, Adamiak M, Deptuła W. Coronins and their role in immunological phenomena. Cent Eur J Immunol 2017; 41:435-441. [PMID: 28450807 PMCID: PMC5382889 DOI: 10.5114/ceji.2016.65143] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 01/06/2016] [Indexed: 02/05/2023] Open
Abstract
Coronins are a large family of proteins occurring in many eukaryotes. In mammals, seven coronin genes have been identified, evidencing that coronins 1 to 6 present classic coronin structure, while coronin 7 is a tandem coronin particle, without a spiral domain, although the best characterised coronin, in terms of both structure and function, is the mammalian coronin 1. It has been proven that they are related to regulation of actin dynamics, e.g. as a result of interaction with the complex of proteins Arp2/3. These proteins also modulate the activity of immune system cells, including lymphocyte T and B cells, neutrophils and macrophages. They are involved in bacterial infections with Mycobacterium tuberculosis, M. leprae and Helicobacter pylori and participate in the response to viral infections, e.g. infections of lymphocytic choriomeningitis virus (LCMV) and vesicular stomatitis Indiana virus (VSV). Also their involvement in autoimmune diseases such as lupus erythematosus has been recorded.
Collapse
Affiliation(s)
| | | | - Mateusz Adamiak
- Department of Immunology, Faculty of Biology, University of Szczecin, Poland
| | - Wiesław Deptuła
- Department of Immunology, Faculty of Biology, University of Szczecin, Poland
| |
Collapse
|
40
|
Glimpse of natural selection of long-lived T-cell clones in healthy life. Proc Natl Acad Sci U S A 2016; 113:9858-63. [PMID: 27535935 DOI: 10.1073/pnas.1601634113] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Homeostatic maintenance of T cells with broad clonal diversity is influenced by both continuing output of young T cells from the thymus and ongoing turnover of preexisting clones in the periphery. In the absence of infection, self and commensal antigens are thought to play important roles in selection and homeostatic maintenance of the T-cell pool. Most naïve T cells are short-lived due to lack of antigen encounter, whereas antigen-experienced T cells may survive and persist as long-lived clones. Thus far, little is known about the homeostasis, antigenic specificity, and clonal diversity of long-lived T-cell clones in peripheral lymphoid organs under healthy living conditions. To identify long-lived T-cell clones in mice, we designed a lineage-tracing method to label a wave of T cells produced in the thymus of young mice. After aging the mice for 1.5 y, we found that lineage-tracked T cells consisted of primarily memory-like T cells and T regulatory cells. T-cell receptor repertoire analysis revealed that the lineage-tracked CD4 memory-like T cells and T regulatory cells exhibited age-dependent enrichment of shared clonotypes. Furthermore, these shared clonotypes were found across different mice maintained in the same housing condition. These findings suggest that nonrandom and shared antigens are involved in controlling selection, retention, and immune tolerance of long-lived T-cell clones under healthy living conditions.
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW As T cells engineered with chimeric antigen receptors (CARs) are entering advanced phases of clinical trial testing with promising results, the potential implications of use in an allogeneic environment are emerging as an important consideration. This review discusses the use of allogeneic CAR therapy, the potential effects of T-cell receptor (TCR) signaling on CAR T-cell efficacy, and the potential for TCR elimination to generate an off-the-shelf product. RECENT FINDINGS The majority of preclinical and clinical data regarding allogeneic T cells are focused on safety of their use given the potential for graft-versus-host disease (GVHD) mediated by the T-cell receptor expressed with the introduced CAR. Recent clinical trials using donor-derived CAR T cells are using either rigorous patient selection or T-cell selection (such as enrichment for virus-specific T cells). Although no GVHD has been reported, the efficacy of the allogeneic CAR treatment needs to be optimized. Several preclinical models limit allogeneic CAR-driven GVHD by utilizing memory T-cell selection, virus-specific T cells, gene-editing techniques, or suicide gene engineering. SUMMARY In the allogeneic environment, the potential effects of TCR signaling on the efficacy of CAR could affect the clinical responses with the use of donor-derived CAR T cells. Better understanding of the functionality of donor-derived T cells for therapy is essential for the development of universal effector cells for CAR therapy.
Collapse
|
42
|
Liu J, Haddad EK, Marceau J, Morabito KM, Rao SS, Filali-Mouhim A, Sekaly RP, Graham BS. A Numerically Subdominant CD8 T Cell Response to Matrix Protein of Respiratory Syncytial Virus Controls Infection with Limited Immunopathology. PLoS Pathog 2016; 12:e1005486. [PMID: 26943673 PMCID: PMC4778879 DOI: 10.1371/journal.ppat.1005486] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 02/10/2016] [Indexed: 11/25/2022] Open
Abstract
CD8 T cells are involved in pathogen clearance and infection-induced pathology in respiratory syncytial virus (RSV) infection. Studying bulk responses masks the contribution of individual CD8 T cell subsets to protective immunity and immunopathology. In particular, the roles of subdominant responses that are potentially beneficial to the host are rarely appreciated when the focus is on magnitude instead of quality of response. Here, by evaluating CD8 T cell responses in CB6F1 hybrid mice, in which multiple epitopes are recognized, we found that a numerically subdominant CD8 T cell response against DbM187 epitope of the virus matrix protein expressed high avidity TCR and enhanced signaling pathways associated with CD8 T cell effector functions. Each DbM187 T effector cell lysed more infected targets on a per cell basis than the numerically dominant KdM282 T cells, and controlled virus replication more efficiently with less pulmonary inflammation and illness than the previously well-characterized KdM282 T cell response. Our data suggest that the clinical outcome of viral infections is determined by the integrated functional properties of a variety of responding CD8 T cells, and that the highest magnitude response may not necessarily be the best in terms of benefit to the host. Understanding how to induce highly efficient and functional T cells would inform strategies for designing vaccines intended to provide T cell-mediated immunity. CD8 T cells play a key role in RSV clearance, immunopathology and disease. Therefore, CD8 T cells can help or harm the host depending on their timing, magnitude, and function. The CD8 T cell response represents a heterogeneous population of cells with phenotypically and functionally diverse subsets, and needs to at least be studied at the level of epitope specificity to understand how to diminish the risk of immunopathology. Studying the bulk response masks distinct contributions of individual CD8 T subsets to immunity and immunopathology. Focusing on CD8 T cell response with the highest magnitude overlooks role of subdominant responses. Here, we studied response to different epitopes and revealed that a numerically subdominant CD8 T cell response against DbM187 epitope of the virus matrix protein controlled virus replication efficiently with limited pulmonary inflammation and illness compared to the previously well-characterized and numerically dominant KdM282 T cell response. Our data show that selectively boosting of epitope-specific CD8 T cell responses may be more beneficial than indiscriminant boosting of all available epitopes to achieve rapid viral clearance while limiting immunopathology. This work has implications for antigen design of vaccines intended to induce T-cell-mediated immunity.
Collapse
Affiliation(s)
- Jie Liu
- Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (JL); (BSG)
| | - Elias K. Haddad
- Drexel University, Division of Infectious Diseases and HIV Medicine, Philadelphia, Pennsylvania, United States of America
| | - Joshua Marceau
- Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Kaitlyn M. Morabito
- Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Srinivas S. Rao
- Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ali Filali-Mouhim
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Rafick-Pierre Sekaly
- Center for AIDS Research, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Barney S. Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail: (JL); (BSG)
| |
Collapse
|
43
|
Control of T cell antigen reactivity via programmed TCR downregulation. Nat Immunol 2016; 17:379-86. [PMID: 26901151 PMCID: PMC4803589 DOI: 10.1038/ni.3386] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 12/22/2015] [Indexed: 12/17/2022]
Abstract
The T cell receptor (TCR) is unique in that its affinity for ligand is unknown prior to encounter and can vary by orders of magnitude. How the immune system regulates individual T cells that display highly different reactivity to antigen remains unclear. Here we identified that activated CD4+ T cells, at the peak of clonal expansion, persistently downregulate TCR expression in proportion to the strength of initial antigen recognition. This programmed response increases the threshold for cytokine production and recall proliferation in a clone-specific manner, ultimately excluding clones with the highest antigen reactivities. Thus, programmed TCR downregulation represents a negative feedback mechanism to constrain T cell effector function with a suitable time delay, thereby allowing pathogen control while avoiding excess inflammatory damage.
Collapse
|
44
|
Thiant S, Moutuou MM, Leboeuf D, Guimond M. Homeostatic cytokines in immune reconstitution and graft-versus-host disease. Cytokine 2016; 82:24-32. [PMID: 26795458 DOI: 10.1016/j.cyto.2016.01.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 01/07/2016] [Accepted: 01/07/2016] [Indexed: 12/15/2022]
Abstract
For numerous patients, allogeneic stem cell transplantation (SCT) is the only therapeutic option that could potentially cure their disease. Despite significant progress made in clinical management of allogeneic SCT, acute graft-versus-host disease (aGVHD) remains the second cause of death after disease recurrence. aGVHD is highly immunosuppressive and the adverse effect of allogeneic SCT on T cell regeneration is typically more important than the levels of immunosuppression normally seen after autologous SCT. In these patients, immune reconstitution often takes several years to occur and restoring immunocompetence after allogeneic SCT represents an important challenge, principally because clinical options are limited and current methods used to accelerate immune reconstitution are associated with increased GVHD. Interleukin-7 and IL-15 are both under clinical investigation and demonstrate the greatest potential on peripheral T cells regeneration in mice and humans. However, awareness has been raised about the use of IL-7 and IL-15 after allogeneic SCT with regards to potential adverse effects on aGVHD. In this review, we will discuss about recent progress made in lymphocyte regeneration, the critical role played by IL-7 and IL-15 in T cell homeostasis and how these cytokines could be used to improve immune reconstitution after allogeneic SCT.
Collapse
Affiliation(s)
- Stéphanie Thiant
- Maisonneuve-Rosemont Research Center, Montreal, Quebec, Canada; Department of Microbiology, Infectiology and Immunology, University of Montreal, Montreal, Quebec, Canada
| | - Moutuaata M Moutuou
- Maisonneuve-Rosemont Research Center, Montreal, Quebec, Canada; Department of Microbiology, Infectiology and Immunology, University of Montreal, Montreal, Quebec, Canada
| | - Dominique Leboeuf
- Department of Microbiology, Infectiology and Immunology, University of Montreal, Montreal, Quebec, Canada
| | - Martin Guimond
- Maisonneuve-Rosemont Research Center, Montreal, Quebec, Canada; Department of Microbiology, Infectiology and Immunology, University of Montreal, Montreal, Quebec, Canada.
| |
Collapse
|
45
|
Yang F, Jin H, Wang J, Sun Q, Yan C, Wei F, Ren X. Adoptive Cellular Therapy (ACT) for Cancer Treatment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 909:169-239. [PMID: 27240459 DOI: 10.1007/978-94-017-7555-7_4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Adoptive cellular therapy (ACT) with various lymphocytes or antigen-presenting cells is one stone in the pillar of cancer immunotherapy, which relies on the tumor-specific T cell. The transfusion of bulk T-cell population into patients is an effective treatment for regression of cancer. In this chapter, we summarize the development of various strategies in ACT for cancer immunotherapy and discuss some of the latest progress and obstacles in technical, safety, and even regulatory aspects to translate these technologies to the clinic. ACT is becoming a potentially powerful approach to cancer treatment. Further experiments and clinical trials are needed to optimize this strategy.
Collapse
Affiliation(s)
- Fan Yang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
| | - Hao Jin
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
| | - Jian Wang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
| | - Qian Sun
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
| | - Cihui Yan
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
| | - Feng Wei
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China
| | - Xiubao Ren
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China.
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China.
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, Huanhuxi Road, Tiyuanbei, Hexi District, Tianjin, 300060, Tianjin, China.
| |
Collapse
|
46
|
Temporal fate mapping reveals age-linked heterogeneity in naive T lymphocytes in mice. Proc Natl Acad Sci U S A 2015; 112:E6917-26. [PMID: 26607449 DOI: 10.1073/pnas.1517246112] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Understanding how our T-cell compartments are maintained requires knowledge of their population dynamics, which are typically quantified over days to weeks using the administration of labels incorporated into the DNA of dividing cells. These studies present snapshots of homeostatic dynamics and have suggested that lymphocyte populations are heterogeneous with respect to rates of division and/or death, although resolving the details of such heterogeneity is problematic. Here we present a method of studying the population dynamics of T cells in mice over timescales of months to years that reveals heterogeneity in rates of division and death with respect to the age of the host at the time of thymic export. We use the transplant conditioning drug busulfan to ablate hematopoetic stem cells in young mice but leave the peripheral lymphocyte compartments intact. Following their reconstitution with congenically labeled (donor) bone marrow, we followed the dilution of peripheral host T cells by donor-derived lymphocytes for a year after treatment. Describing these kinetics with mathematical models, we estimate rates of thymic production, division and death of naive CD4 and CD8 T cells. Population-averaged estimates of mean lifetimes are consistent with earlier studies, but we find the strongest support for a model in which both naive T-cell pools contain kinetically distinct subpopulations of older host-derived cells with self-renewing capacity that are resistant to displacement by naive donor lymphocytes. We speculate that these incumbent cells are conditioned or selected for increased fitness through homeostatic expansion into the lymphopenic neonatal environment.
Collapse
|
47
|
Lythe G, Callard RE, Hoare RL, Molina-París C. How many TCR clonotypes does a body maintain? J Theor Biol 2015; 389:214-24. [PMID: 26546971 PMCID: PMC4678146 DOI: 10.1016/j.jtbi.2015.10.016] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 09/13/2015] [Accepted: 10/07/2015] [Indexed: 01/08/2023]
Abstract
We consider the lifetime of a T cell clonotype, the set of T cells with the same T cell receptor, from its thymic origin to its extinction in a multiclonal repertoire. Using published estimates of total cell numbers and thymic production rates, we calculate the mean number of cells per TCR clonotype, and the total number of clonotypes, in mice and humans. When there is little peripheral division, as in a mouse, the number of cells per clonotype is small and governed by the number of cells with identical TCR that exit the thymus. In humans, peripheral division is important and a clonotype may survive for decades, during which it expands to comprise many cells. We therefore devise and analyse a computational model of homeostasis of a multiclonal population. Each T cell in the model competes for self pMHC stimuli, cells of any one clonotype only recognising a small fraction of the many subsets of stimuli. A constant mean total number of cells is maintained by a balance between cell division and death, and a stable number of clonotypes by a balance between thymic production of new clonotypes and extinction of existing ones. The number of distinct clonotypes in a human body may be smaller than the total number of naive T cells by only one order of magnitude.
The number of T cells of one clonotype is an integer. The history of a clonotype starts with release from the thymus, and ends with extinction. Competition and cross-reactivity are included in a natural way. The average number of cells per clonotype, in a human body, is only of order 10.
Collapse
Affiliation(s)
- Grant Lythe
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds LS2 9JT, UK.
| | - Robin E Callard
- Institute for Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK; Centre for Mathematics and Physics in the Life Sciences and Experimental Biology, University College London, Gower Street, London WC1N 1EH, UK
| | - Rollo L Hoare
- Institute for Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK; Centre for Mathematics and Physics in the Life Sciences and Experimental Biology, University College London, Gower Street, London WC1N 1EH, UK
| | - Carmen Molina-París
- Department of Applied Mathematics, School of Mathematics, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
48
|
Gauthier SD, Leboeuf D, Manuguerra-Gagné R, Gaboury L, Guimond M. Stromal-Derived Factor-1α and Interleukin-7 Treatment Improves Homeostatic Proliferation of Naïve CD4(+) T Cells after Allogeneic Stem Cell Transplantation. Biol Blood Marrow Transplant 2015; 21:1721-31. [PMID: 26151303 DOI: 10.1016/j.bbmt.2015.06.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 06/30/2015] [Indexed: 11/19/2022]
Abstract
Graft-versus-host disease (GVHD) impairs immune reconstitution after allogeneic stem cell transplantation (allo-SCT) and effective therapies aimed at restoring T cell counts in GVHD patients have yet to be developed. During GVHD, CD4(+) T cell reconstitution is particularly affected and current models hold that GVHD insult to the peripheral lymphoid niche is responsible for this effect. Here, we show that naïve CD4(+) T cell homeostatic proliferation (HP) is lost during GVHD because of low systemic IL-7 and impaired dendritic cell (DC) regeneration. We assessed factors involved in DC differentiation and found that although fms-like tyrosine kinase 3 ligand (Flt3-L) levels were normal, stromal-derived factor-1α (SDF-1α) was diminished in the blood of GVHD mice. Unlike Flt3-L treatment, the administration of SDF-1α specifically increased CD8α(+) DC numbers and did not worsen GVHD. Importantly, CD4(+) T cell HP was enhanced only when IL-7 and SDF-1α or Flt3L were coadministered, confirming the crucial role of DCs and IL-7 in restoring CD4(+) T cell regeneration during GVHD. Altogether, our results indicate that CD8α(+) DCs are part of the peripheral niche that controls CD4(+) T cell HP and that their depletion, combined with low systemic IL-7, explains how GVHD constrains naïve CD4(+) T cell reconstitution after allo-SCT.
Collapse
Affiliation(s)
- Simon-David Gauthier
- Department of Hematology-Oncology, Maisonneuve-Rosemont Hospital Research Center, Montréal, Québec, Canada; Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Dominique Leboeuf
- Department of Hematology-Oncology, Maisonneuve-Rosemont Hospital Research Center, Montréal, Québec, Canada
| | - Renaud Manuguerra-Gagné
- Department of Hematology-Oncology, Maisonneuve-Rosemont Hospital Research Center, Montréal, Québec, Canada
| | - Louis Gaboury
- Department of Pathology, Institute for Research in Immunology and Cancer, Montréal, Québec, Canada
| | - Martin Guimond
- Department of Hematology-Oncology, Maisonneuve-Rosemont Hospital Research Center, Montréal, Québec, Canada; Department of Microbiology, Infectiology and Immunology, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
49
|
Catalán E, Charni S, Jaime P, Aguiló JI, Enríquez JA, Naval J, Pardo J, Villalba M, Anel A. MHC-I modulation due to changes in tumor cell metabolism regulates tumor sensitivity to CTL and NK cells. Oncoimmunology 2015; 4:e985924. [PMID: 25949869 DOI: 10.4161/2162402x.2014.985924] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 11/05/2014] [Indexed: 12/19/2022] Open
Abstract
Tumor cells have a tendency to use glucose fermentation to obtain energy instead of mitochondrial oxidative phosphorylation (OXPHOS). We demonstrated that this phenotype correlated with loss of ERK5 expression and with reduced MHC class I expression. Consequently, tumor cells could evade cytotoxic T lymphocyte (CTL)-mediated immune surveillance, but also increase their sensitivity to natural killer (NK) cells. These outcomes were evaluated using two cellular models: leukemic EL4 cells and L929 transformed fibroblasts and their derived ρ° cell lines, which lack mitochondrial DNA. We have also used a L929 cell sub-line that spontaneously lost matrix attachment (L929dt), reminiscent of metastasis generation, that also downregulated MHC-I and ERK5 expression. MHC-I expression is lower in ρ° cells than in the parental cell lines, but they were equally sensitive to CTL. On the contrary, ρ° cells were more sensitive to activated NK cells than parental cells. On the other hand, L929dt cells were resistant to CTL and NK cells, showed reduced viability when forced to perform OXPHOS, and surviving cells increased MHC-I expression and became sensitive to CTL. The present results suggest that when the reduction in MHC-I levels in tumor cells due to glycolytic metabolism is partial, the increase in sensitivity to NK cells seems to predominate. However, when tumor cells completely lose MHC-I expression, the combination of treatments that increase OXPHOS with CTL-mediated immunotherapy could be a promising therapeutic approach.
Collapse
Affiliation(s)
- Elena Catalán
- Apoptosis; Immunity & Cancer Group; Dept. Biochemistry and Molecular and Cell Biology; Faculty of Sciences; Campus San Francisco Sq.; University of Zaragoza and Aragón Health Research Institute (IIS Aragón) ; Zaragoza, Spain
| | - Seyma Charni
- INSERM-UM1 U1040; Université de Montpellier 1,UFR Médecine ; Montpellier, France ; Institut de Recherche en Biothérapie (IRB); CHU Montpellier ; Hôpital Saint-Eloi, 80, Av. Augustin Fliche ; Montpellier, France
| | - Paula Jaime
- Immune Effector Cells Group; IIS Aragón; Biomedical Research Centre of Aragón (CIBA)-Nanoscience Institute of Aragon (INA) ; Avda. San Juan Bosco ; Zaragoza, Spain
| | - Juan Ignacio Aguiló
- Apoptosis; Immunity & Cancer Group; Dept. Biochemistry and Molecular and Cell Biology; Faculty of Sciences; Campus San Francisco Sq.; University of Zaragoza and Aragón Health Research Institute (IIS Aragón) ; Zaragoza, Spain
| | - José Antonio Enríquez
- Dept. Biochemistry and Molecular and Cell Biology; University of Zaragoza and Dept. of Cardiovascular Development and Repair; National Center for Cardiovascular Research Carlos III; Melchor Fernandez Almagro ; Madrid, Spain
| | - Javier Naval
- Apoptosis; Immunity & Cancer Group; Dept. Biochemistry and Molecular and Cell Biology; Faculty of Sciences; Campus San Francisco Sq.; University of Zaragoza and Aragón Health Research Institute (IIS Aragón) ; Zaragoza, Spain
| | - Julián Pardo
- Immune Effector Cells Group; IIS Aragón; Biomedical Research Centre of Aragón (CIBA)-Nanoscience Institute of Aragon (INA) ; Avda. San Juan Bosco ; Zaragoza, Spain ; Aragón I+D Foundation (ARAID) ; Avda. San Juan Bosco ; Zaragoza, Spain
| | - Martín Villalba
- INSERM-UM1 U1040; Université de Montpellier 1,UFR Médecine ; Montpellier, France ; Institut de Recherche en Biothérapie (IRB); CHU Montpellier ; Hôpital Saint-Eloi, 80, Av. Augustin Fliche ; Montpellier, France
| | - Alberto Anel
- Apoptosis; Immunity & Cancer Group; Dept. Biochemistry and Molecular and Cell Biology; Faculty of Sciences; Campus San Francisco Sq.; University of Zaragoza and Aragón Health Research Institute (IIS Aragón) ; Zaragoza, Spain
| |
Collapse
|
50
|
Blanco R, Borroto A, Schamel W, Pereira P, Alarcon B. Conformational changes in the T cell receptor differentially determine T cell subset development in mice. Sci Signal 2014; 7:ra115. [DOI: 10.1126/scisignal.2005650] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|