1
|
Bürdek M, Prinz PU, Mutze K, Tippmer S, Geiger C, Longinotti G, Schendel DJ. Characterization of a 3S PRAME VLD-Specific T Cell Receptor and Its Use in Investigational Medicinal Products for TCR-T Therapy of Patients with Myeloid Malignancies. Cancers (Basel) 2025; 17:242. [PMID: 39858024 PMCID: PMC11763942 DOI: 10.3390/cancers17020242] [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: 11/12/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES MDG1011 is an autologous TCR-T therapy developed as a treatment option for patients with myeloid malignancies, including acute myeloid leukemia (AML), myelodysplastic syndrome (MDS), and multiple myeloma (MM). It is specific for the target antigen PReferentially expressed Antigen in MElanoma (PRAME). The recombinant TCR used in MDG1011 recognizes PRAME100-108 VLD-peptide presented by HLA-A*02:01-encoded surface molecules. METHODS Two preclinical batches of MDG1011, produced from enriched CD8+ T cells of healthy donors, underwent rigorous evaluation of on-target and off-target recognition of tumor cells and test cells representing healthy tissues. MDG1011 investigational medicinal products (IMPs) were produced for 13 patients. VLD-TCR surface expression was assessed using dual-marker flow cytometry using TCR V-beta-specific antibody and VLD/HLA-A2-specific multimer. Functionality was assessed by interferon-gamma (IFN-γ) secretion and cell-mediated cytotoxicity of target cells. RESULTS Preclinical MDG1011 batches displayed strong VLD-TCR expression, cytokine secretion, and cytotoxicity after antigen-specific activation, while showing no signals of on-target/off-tumor or off-target recognition. All IMPs had good VLD-TCR expression as well as functionality after activation by multiple target cells. CONCLUSIONS Preclinical studies demonstrated that MDG1011 displayed key 3S attributes of high specificity, sensitivity, and safety required for regulatory approval of a first-in-human (FIH) clinical study of patients with myeloid malignancies (CD-TCR-001: ClinicalTrials.gov Identifier: NCT03503968). MDG1011 IMP manufacturing was successful at 92%, even including heavily pretreated elderly patients with very advanced disease. The IMPs applied in nine patients all displayed antigen-specific functionality. Elsewhere, clinical study results for MDG1011 showed no dose-limiting toxicity and signs of biological and/or clinical activity in several patients.
Collapse
Affiliation(s)
- Maja Bürdek
- Medigene Immunotherapies GmbH, 82152 Planegg-Martinsried, Germany; (P.U.P.); (K.M.); (S.T.); (C.G.); (G.L.)
| | - Petra U. Prinz
- Medigene Immunotherapies GmbH, 82152 Planegg-Martinsried, Germany; (P.U.P.); (K.M.); (S.T.); (C.G.); (G.L.)
| | - Kathrin Mutze
- Medigene Immunotherapies GmbH, 82152 Planegg-Martinsried, Germany; (P.U.P.); (K.M.); (S.T.); (C.G.); (G.L.)
| | - Stefanie Tippmer
- Medigene Immunotherapies GmbH, 82152 Planegg-Martinsried, Germany; (P.U.P.); (K.M.); (S.T.); (C.G.); (G.L.)
| | - Christiane Geiger
- Medigene Immunotherapies GmbH, 82152 Planegg-Martinsried, Germany; (P.U.P.); (K.M.); (S.T.); (C.G.); (G.L.)
| | - Giulia Longinotti
- Medigene Immunotherapies GmbH, 82152 Planegg-Martinsried, Germany; (P.U.P.); (K.M.); (S.T.); (C.G.); (G.L.)
| | - Dolores J. Schendel
- Medigene Immunotherapies GmbH, 82152 Planegg-Martinsried, Germany; (P.U.P.); (K.M.); (S.T.); (C.G.); (G.L.)
- Medigene AG, 82152 Planegg-Martinsried, Germany
| |
Collapse
|
2
|
Chai T, Loh KM, Weissman IL. TMX1, a disulfide oxidoreductase, is necessary for T cell function through regulation of CD3ζ. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.22.614388. [PMID: 39386445 PMCID: PMC11463681 DOI: 10.1101/2024.09.22.614388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
T cell-targeted therapies are commonly used to manage T cell hyperactivity in autoimmune disorders, graft-versus-host diseases (GVHD), and transplant rejections. However, many patients experience significant side effects or inadequate responses to current treatments, highlighting the urgent need for alternative strategies. In this study, we searched for regulators of T cells through proximity labeling with APEX2 to detect proteins interacting with CD8α, a coreceptor of the T-cell receptor (TCR). This screen revealed TMX1, an ER resident transmembrane disulfide oxidoreductase, is essential for T cell cytotoxicity and NFAT, NFκB, and AP1 signaling but not cell proliferation. TMX1 deletion decreases surface TCR expression and destabilizes CD3ζ, a subunit of TCR complex; however, overexpression of CD3ζ rescues the phenotype, suggesting that TMX1 is not required for CD3ζ function. Mechanistically, TMX1 was found to directly engage the CxxC motif of CD3δ, which has been reported to be essential for proper TCR assembly and function. We hypothesize that the loss of TMX1 interaction with CD3δ leads to impaired TCR assembly and subsequent CD3ζ destabilization. These findings identify TMX1 as a novel regulator of T-cell receptor assembly and a potential target for immunosuppressive therapy.
Collapse
Affiliation(s)
- Timothy Chai
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Kyle M. Loh
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Irving L. Weissman
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
3
|
Hong H, Wang Y, Menard M, Buckley JA, Zhou L, Volpicelli-Daley L, Standaert DG, Qin H, Benveniste EN. Suppression of the JAK/STAT pathway inhibits neuroinflammation in the line 61-PFF mouse model of Parkinson's disease. J Neuroinflammation 2024; 21:216. [PMID: 39218899 PMCID: PMC11368013 DOI: 10.1186/s12974-024-03210-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
Parkinson's disease (PD) is characterized by neuroinflammation, progressive loss of dopaminergic neurons, and accumulation of α-synuclein (α-Syn) into insoluble aggregates called Lewy pathology. The Line 61 α-Syn mouse is an established preclinical model of PD; Thy-1 is used to promote human α-Syn expression, and features of sporadic PD develop at 9-18 months of age. To accelerate the PD phenotypes, we injected sonicated human α-Syn preformed fibrils (PFFs) into the striatum, which produced phospho-Syn (p-α-Syn) inclusions in the substantia nigra pars compacta and significantly increased MHC Class II-positive immune cells. Additionally, there was enhanced infiltration and activation of innate and adaptive immune cells in the midbrain. We then used this new model, Line 61-PFF, to investigate the effect of inhibiting the JAK/STAT signaling pathway, which is critical for regulation of innate and adaptive immune responses. After administration of the JAK1/2 inhibitor AZD1480, immunofluorescence staining showed a significant decrease in p-α-Syn inclusions and MHC Class II expression. Flow cytometry showed reduced infiltration of CD4+ T-cells, CD8+ T-cells, CD19+ B-cells, dendritic cells, macrophages, and endogenous microglia into the midbrain. Importantly, single-cell RNA-Sequencing analysis of CD45+ cells from the midbrain identified 9 microglia clusters, 5 monocyte/macrophage (MM) clusters, and 5 T-cell (T) clusters, in which potentially pathogenic MM4 and T3 clusters were associated with neuroinflammatory responses in Line 61-PFF mice. AZD1480 treatment reduced cell numbers and cluster-specific expression of the antigen-presentation genes H2-Eb1, H2-Aa, H2-Ab1, and Cd74 in the MM4 cluster and proinflammatory genes such as Tnf, Il1b, C1qa, and C1qc in the T3 cluster. Together, these results indicate that inhibiting the JAK/STAT pathway suppresses the activation and infiltration of innate and adaptive cells, reducing neuroinflammation in the Line 61-PFF mouse model.
Collapse
Affiliation(s)
- Huixian Hong
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, 1918 University Boulevard, MCLM 907, Birmingham, AL, 35294, USA
| | - Yong Wang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, 1918 University Boulevard, MCLM 907, Birmingham, AL, 35294, USA
| | - Marissa Menard
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Jessica A Buckley
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, 1918 University Boulevard, MCLM 907, Birmingham, AL, 35294, USA
| | - Lianna Zhou
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, 1918 University Boulevard, MCLM 907, Birmingham, AL, 35294, USA
| | - Laura Volpicelli-Daley
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - David G Standaert
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Hongwei Qin
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, 1918 University Boulevard, MCLM 907, Birmingham, AL, 35294, USA.
| | - Etty N Benveniste
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, 1918 University Boulevard, MCLM 907, Birmingham, AL, 35294, USA.
| |
Collapse
|
4
|
Hong H, Wang Y, Menard M, Buckley J, Zhou L, Volpicelli-Daley L, Standaert D, Qin H, Benveniste E. Suppression of the JAK/STAT Pathway Inhibits Neuroinflammation in the Line 61-PFF Mouse Model of Parkinson's Disease. RESEARCH SQUARE 2024:rs.3.rs-4307273. [PMID: 38766241 PMCID: PMC11100885 DOI: 10.21203/rs.3.rs-4307273/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Parkinson's disease (PD) is characterized by neuroinflammation, progressive loss of dopaminergic neurons, and accumulation of a-synuclein (a-Syn) into insoluble aggregates called Lewy pathology. The Line 61 a-Syn mouse is an established preclinical model of PD; Thy-1 is used to promote human a-Syn expression, and features of sporadic PD develop at 9-18 months of age. To accelerate the PD phenotypes, we injected sonicated human a-Syn preformed fibrils (PFFs) into the striatum, which produced phospho-Syn (p-a-Syn) inclusions in the substantia nigra pars compacta and significantly increased MHC Class II-positive immune cells. Additionally, there was enhanced infiltration and activation of innate and adaptive immune cells in the midbrain. We then used this new model, Line 61-PFF, to investigate the effect of inhibiting the JAK/STAT signaling pathway, which is critical for regulation of innate and adaptive immune responses. After administration of the JAK1/2 inhibitor AZD1480, immunofluorescence staining showed a significant decrease in p-a-Syn inclusions and MHC Class II expression. Flow cytometry showed reduced infiltration of CD4+ T-cells, CD8+ T-cells, CD19+ B-cells, dendritic cells, macrophages, and endogenous microglia into the midbrain. Importantly, single-cell RNA-Sequencing analysis of CD45+ cells from the midbrain identified 9 microglia clusters, 5 monocyte/macrophage (MM) clusters, and 5 T-cell (T) clusters, in which potentially pathogenic MM4 and T3 clusters were associated with neuroinflammatory responses in Line 61-PFF mice. AZD1480 treatment reduced cell numbers and cluster-specific expression of the antigen-presentation genes H2-Eb1, H2-Aa, H2-Ab1, and Cd74 in the MM4 cluster and proinflammatory genes such as Tnf, Il1b, C1qa, and C1qc in the T3 cluster. Together, these results indicate that inhibiting the JAK/STAT pathway suppresses the activation and infiltration of innate and adaptive cells, reducing neuroinflammation in the Line 61-PFF mouse model.
Collapse
|
5
|
Sattler A, Gamradt S, Proß V, Thole LML, He A, Schrezenmeier EV, Jechow K, Gold SM, Lukassen S, Conrad C, Kotsch K. CD3 downregulation identifies high-avidity, multipotent SARS-CoV-2 vaccine- and recall antigen-specific Th cells with distinct metabolism. JCI Insight 2024; 9:e166833. [PMID: 38206757 PMCID: PMC11143931 DOI: 10.1172/jci.insight.166833] [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: 11/02/2022] [Accepted: 01/09/2024] [Indexed: 01/13/2024] Open
Abstract
Functional avidity is supposed to critically shape the quality of immune responses, thereby influencing host protection against infectious agents including SARS-CoV-2. Here we show that after human SARS-CoV-2 vaccination, a large portion of high-avidity spike-specific CD4+ T cells lost CD3 expression after in vitro activation. The CD3- subset was enriched for cytokine-positive cells, including elevated per-cell expression levels, and showed increased polyfunctionality. Assessment of key metabolic pathways by flow cytometry revealed that superior functionality was accompanied by a shift toward fatty acid synthesis at the expense of their oxidation, whereas glucose transport and glycolysis were similarly regulated in SARS-CoV-2-specific CD3- and CD3+ subsets. As opposed to their CD3+ counterparts, frequencies of vaccine-specific CD3- T cells positively correlated with both the size of the naive CD4+ T cell pool and vaccine-specific IgG levels. Moreover, their frequencies negatively correlated with advancing age and were impaired in patients under immunosuppressive therapy. Typical recall antigen-reactive T cells showed a comparable segregation into functionally and metabolically distinct CD3+ and CD3- subsets but were quantitatively maintained upon aging, likely due to earlier recruitment in life. In summary, our data identify CD3- T helper cells as correlates of high-quality immune responses that are impaired in at-risk populations.
Collapse
Affiliation(s)
- Arne Sattler
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department for General and Visceral Surgery, Berlin, Germany
| | - Stefanie Gamradt
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Neurosciences – Campus Benjamin Franklin, Berlin, Germany
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychosomatic Medicine – Campus Benjamin Franklin, Berlin, Germany
| | - Vanessa Proß
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department for General and Visceral Surgery, Berlin, Germany
| | - Linda Marie Laura Thole
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department for General and Visceral Surgery, Berlin, Germany
| | - An He
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department for General and Visceral Surgery, Berlin, Germany
| | - Eva Vanessa Schrezenmeier
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Nephrology and Medical Intensive Care, Berlin, Germany
| | - Katharina Jechow
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Center for Digital Health, Berlin, Germany
| | - Stefan M. Gold
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychiatry and Neurosciences – Campus Benjamin Franklin, Berlin, Germany
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Psychosomatic Medicine – Campus Benjamin Franklin, Berlin, Germany
- Universitätsklinikum Hamburg Eppendorf, Institut für Neuroimmunologie und Multiple Sklerose, Hamburg, Germany
| | - Sören Lukassen
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Center for Digital Health, Berlin, Germany
| | - Christian Conrad
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Center for Digital Health, Berlin, Germany
| | - Katja Kotsch
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department for General and Visceral Surgery, Berlin, Germany
| |
Collapse
|
6
|
Ebbinghaus M, Wittich K, Bancher B, Lebedeva V, Appelshoffer A, Femel J, Helm MS, Kollet J, Hardt O, Pfeifer R. Endogenous Signaling Molecule Activating (ESMA) CARs: A Novel CAR Design Showing a Favorable Risk to Potency Ratio for the Treatment of Triple Negative Breast Cancer. Int J Mol Sci 2024; 25:615. [PMID: 38203786 PMCID: PMC10779313 DOI: 10.3390/ijms25010615] [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: 11/30/2023] [Revised: 12/19/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
As chimeric antigen receptor (CAR) T cell therapy continues to gain attention as a valuable treatment option against different cancers, strategies to improve its potency and decrease the side effects associated with this therapy have become increasingly relevant. Herein, we report an alternative CAR design that incorporates transmembrane domains with the ability to recruit endogenous signaling molecules, eliminating the need for stimulatory signals within the CAR structure. These endogenous signaling molecule activating (ESMA) CARs triggered robust cytotoxic activity and proliferation of the T cells when directed against the triple-negative breast cancer (TNBC) cell line MDA-MB-231 while exhibiting reduced cytokine secretion and exhaustion marker expression compared to their cognate standard second generation CARs. In a NOD SCID Gamma (NSG) MDA-MB-231 xenograft mouse model, the lead candidate maintained longitudinal therapeutic efficacy and an enhanced T cell memory phenotype. Profound tumor infiltration by activated T cells repressed tumor growth, further manifesting the proliferative capacity of the ESMA CAR T cell therapy. Consequently, ESMA CAR T cells entail promising features for improved clinical outcome as a solid tumor treatment option.
Collapse
Affiliation(s)
- Mira Ebbinghaus
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany; (M.E.); (K.W.); (B.B.); (V.L.); (A.A.); (J.F.); (M.S.H.); (J.K.)
- School of Applied Biosciences and Chemistry, HAN University of Applied Sciences, 6525 EM Nijmegen, The Netherlands
| | - Katharina Wittich
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany; (M.E.); (K.W.); (B.B.); (V.L.); (A.A.); (J.F.); (M.S.H.); (J.K.)
| | - Benjamin Bancher
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany; (M.E.); (K.W.); (B.B.); (V.L.); (A.A.); (J.F.); (M.S.H.); (J.K.)
| | - Valeriia Lebedeva
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany; (M.E.); (K.W.); (B.B.); (V.L.); (A.A.); (J.F.); (M.S.H.); (J.K.)
| | - Anijutta Appelshoffer
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany; (M.E.); (K.W.); (B.B.); (V.L.); (A.A.); (J.F.); (M.S.H.); (J.K.)
| | - Julia Femel
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany; (M.E.); (K.W.); (B.B.); (V.L.); (A.A.); (J.F.); (M.S.H.); (J.K.)
| | - Martin S. Helm
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany; (M.E.); (K.W.); (B.B.); (V.L.); (A.A.); (J.F.); (M.S.H.); (J.K.)
| | - Jutta Kollet
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany; (M.E.); (K.W.); (B.B.); (V.L.); (A.A.); (J.F.); (M.S.H.); (J.K.)
| | - Olaf Hardt
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany; (M.E.); (K.W.); (B.B.); (V.L.); (A.A.); (J.F.); (M.S.H.); (J.K.)
| | - Rita Pfeifer
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany; (M.E.); (K.W.); (B.B.); (V.L.); (A.A.); (J.F.); (M.S.H.); (J.K.)
| |
Collapse
|
7
|
Kalinina AA, Khromykh LM, Kazansky DB. T Cell Receptor Chain Centricity: The Phenomenon and Potential Applications in Cancer Immunotherapy. Int J Mol Sci 2023; 24:15211. [PMID: 37894892 PMCID: PMC10607890 DOI: 10.3390/ijms242015211] [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: 08/31/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 10/29/2023] Open
Abstract
T cells are crucial players in adaptive anti-cancer immunity. The gene modification of T cells with tumor antigen-specific T cell receptors (TCRs) was a milestone in personalized cancer immunotherapy. TCR is a heterodimer (either α/β or γ/δ) able to recognize a peptide antigen in a complex with self-MHC molecules. Although traditional concepts assume that an α- and β-chain contribute equally to antigen recognition, mounting data reveal that certain receptors possess chain centricity, i.e., one hemi-chain TCR dominates antigen recognition and dictates its specificity. Chain-centric TCRs are currently poorly understood in terms of their origin and the functional T cell subsets that express them. In addition, the ratio of α- and β-chain-centric TCRs, as well as the exact proportion of chain-centric TCRs in the native repertoire, is generally still unknown today. In this review, we provide a retrospective analysis of studies that evidence chain-centric TCRs, propose patterns of their generation, and discuss the potential applications of such receptors in T cell gene modification for adoptive cancer immunotherapy.
Collapse
Affiliation(s)
| | | | - Dmitry B. Kazansky
- N.N. Blokhin National Medical Research Center of Oncology of the Ministry of Health of the Russian Federation, 115478 Moscow, Russia
| |
Collapse
|
8
|
Dustin ML. Recent advances in understanding TCR signaling: a synaptic perspective. Fac Rev 2023; 12:25. [PMID: 37900153 PMCID: PMC10608137 DOI: 10.12703/r/12-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2023] Open
Abstract
The T cell receptor is a multi-subunit complex that carries out a range of recognition tasks for multiple lymphocyte types and translates recognition into signals that regulate survival, growth, differentiation, and effector functions for innate and adaptive host defense. Recent advances include the cryo-electron microscopy-based structure of the extracellular and transmembrane components of the complex, new information about coupling to intracellular partners, lateral associations in the membrane that all add to our picture of the T cell signaling machinery, and how signal termination relates to effector function. This review endeavors to integrate structural and biochemical information through the lens of the immunological synapse- the critical interface with the antigen-presenting cell.
Collapse
Affiliation(s)
- Michael L Dustin
- Kennedy Institute of Rheumatology, The University of Oxford, Oxford, UK
| |
Collapse
|
9
|
Giacalone VD, Giraldo DM, Silva GL, Hosten J, Peng L, Guglani L, Tirouvanziam R. Pulmonary exacerbations in early cystic fibrosis lung disease are marked by strong modulation of CD3 and PD-1 on luminal T cells. Front Immunol 2023; 14:1194253. [PMID: 37809107 PMCID: PMC10551126 DOI: 10.3389/fimmu.2023.1194253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
Background In chronic cystic fibrosis (CF) lung disease, neutrophilic inflammation and T-cell inhibition occur concomitantly, partly due to neutrophil-mediated release of the T-cell inhibitory enzyme Arg1. However, the onset of this tonic inhibition of T cells, and the impact of pulmonary exacerbations (PEs) on this process, remain unknown. Methods Children with CF aged 0-5 years were enrolled in a longitudinal, single-center cohort study. Blood (n = 35) and bronchoalveolar lavage (BAL) fluid (n = 18) were collected at stable outpatient clinic visits or inpatient PE hospitalizations and analyzed by flow cytometry (for immune cell presence and phenotype) and 20-plex chemiluminescence assay (for immune mediators). Patients were categorized by PE history into (i) no prior PE, (ii) past history of PE prior to stable visit, or (iii) current PE. Results PEs were associated with increased concentration of both pro- and anti-inflammatory mediators in BAL, and increased neutrophil frequency and G-CSF in circulation. PE BAL samples showed a trend toward an increased frequency of hyperexocytic "GRIM" neutrophils, which we previously identified in chronic CF. Interestingly, expression levels of the T-cell receptor associated molecule CD3 and of the inhibitory programmed death-1 (PD-1) receptor were respectively decreased and increased on T cells from BAL compared to blood in all patients. When categorized by PE status, CD3 and PD-1 expression on blood T cells did not differ among patients, while CD3 expression was decreased, and PD-1 expression was increased on BAL T cells from patients with current PE. Conclusions Our findings suggest that airway T cells are engaged during early-life PEs, prior to the onset of chronic neutrophilic inflammation in CF. In addition, increased blood neutrophil frequency and a trend toward increased BAL frequency of hyperexocytic neutrophils suggest that childhood PEs may progressively shift the balance of CF airway immunity towards neutrophil dominance.
Collapse
Affiliation(s)
- Vincent D. Giacalone
- Department of Pediatrics, Emory University, Atlanta, GA, United States
- Center for CF and Airways Disease Research, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Diego Moncada Giraldo
- Department of Pediatrics, Emory University, Atlanta, GA, United States
- Center for CF and Airways Disease Research, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - George L. Silva
- Department of Pediatrics, Emory University, Atlanta, GA, United States
- Center for CF and Airways Disease Research, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Justin Hosten
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Limin Peng
- Department of Biostatistics and Bioinformatics, Emory University School of Public Health, Atlanta, GA, United States
| | - Lokesh Guglani
- Department of Pediatrics, Emory University, Atlanta, GA, United States
- Center for CF and Airways Disease Research, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Rabindra Tirouvanziam
- Department of Pediatrics, Emory University, Atlanta, GA, United States
- Center for CF and Airways Disease Research, Children’s Healthcare of Atlanta, Atlanta, GA, United States
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| |
Collapse
|
10
|
Schendel DJ. Evolution by innovation as a driving force to improve TCR-T therapies. Front Oncol 2023; 13:1216829. [PMID: 37810959 PMCID: PMC10552759 DOI: 10.3389/fonc.2023.1216829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/16/2023] [Indexed: 10/10/2023] Open
Abstract
Adoptive cell therapies continually evolve through science-based innovation. Specialized innovations for TCR-T therapies are described here that are embedded in an End-to-End Platform for TCR-T Therapy Development which aims to provide solutions for key unmet patient needs by addressing challenges of TCR-T therapy, including selection of target antigens and suitable T cell receptors, generation of TCR-T therapies that provide long term, durable efficacy and safety and development of efficient and scalable production of patient-specific (personalized) TCR-T therapy for solid tumors. Multiple, combinable, innovative technologies are used in a systematic and sequential manner in the development of TCR-T therapies. One group of technologies encompasses product enhancements that enable TCR-T therapies to be safer, more specific and more effective. The second group of technologies addresses development optimization that supports discovery and development processes for TCR-T therapies to be performed more quickly, with higher quality and greater efficiency. Each module incorporates innovations layered onto basic technologies common to the field of immunology. An active approach of "evolution by innovation" supports the overall goal to develop best-in-class TCR-T therapies for treatment of patients with solid cancer.
Collapse
Affiliation(s)
- Dolores J. Schendel
- Medigene Immunotherapies GmbH, Planegg, Germany
- Medigene AG, Planegg, Germany
| |
Collapse
|
11
|
Rodríguez-Galán A, Dosil SG, Hrčková A, Fernández-Messina L, Feketová Z, Pokorná J, Fernández-Delgado I, Camafeita E, Gómez MJ, Ramírez-Huesca M, Gutiérrez-Vázquez C, Sánchez-Cabo F, Vázquez J, Vaňáčová Š, Sánchez-Madrid F. ISG20L2: an RNA nuclease regulating T cell activation. Cell Mol Life Sci 2023; 80:273. [PMID: 37646974 PMCID: PMC10468436 DOI: 10.1007/s00018-023-04925-2] [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: 04/12/2023] [Revised: 08/07/2023] [Accepted: 08/16/2023] [Indexed: 09/01/2023]
Abstract
ISG20L2, a 3' to 5' exoribonuclease previously associated with ribosome biogenesis, is identified here in activated T cells as an enzyme with a preferential affinity for uridylated miRNA substrates. This enzyme is upregulated in T lymphocytes upon TCR and IFN type I stimulation and appears to be involved in regulating T cell function. ISG20L2 silencing leads to an increased basal expression of CD69 and induces greater IL2 secretion. However, ISG20L2 absence impairs CD25 upregulation, CD3 synaptic accumulation and MTOC translocation towards the antigen-presenting cell during immune synapsis. Remarkably, ISG20L2 controls the expression of immunoregulatory molecules, such as AHR, NKG2D, CTLA-4, CD137, TIM-3, PD-L1 or PD-1, which show increased levels in ISG20L2 knockout T cells. The dysregulation observed in these key molecules for T cell responses support a role for this exonuclease as a novel RNA-based regulator of T cell function.
Collapse
Affiliation(s)
- Ana Rodríguez-Galán
- Instituto Investigación Sanitaria Princesa (IIS-IP), Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Intercellular Communication in the Inflammatory Response, Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Universidad Autónoma de Madrid, Madrid, Spain
| | - Sara G Dosil
- Instituto Investigación Sanitaria Princesa (IIS-IP), Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Intercellular Communication in the Inflammatory Response, Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Universidad Autónoma de Madrid, Madrid, Spain
| | - Anna Hrčková
- CEITEC-Central European Institute of Technology, Masaryk University, Kamenice 5/A35, 625 00, Brno, Czech Republic
- Faculty of Medicine, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
| | - Lola Fernández-Messina
- Instituto Investigación Sanitaria Princesa (IIS-IP), Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Intercellular Communication in the Inflammatory Response, Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red, Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Universidad Complutense de Madrid, Madrid, Spain
| | - Zuzana Feketová
- CEITEC-Central European Institute of Technology, Masaryk University, Kamenice 5/A35, 625 00, Brno, Czech Republic
| | - Julie Pokorná
- CEITEC-Central European Institute of Technology, Masaryk University, Kamenice 5/A35, 625 00, Brno, Czech Republic
| | - Irene Fernández-Delgado
- Instituto Investigación Sanitaria Princesa (IIS-IP), Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Intercellular Communication in the Inflammatory Response, Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Universidad Autónoma de Madrid, Madrid, Spain
| | - Emilio Camafeita
- Centro de Investigación Biomédica en Red, Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Manuel José Gómez
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Marta Ramírez-Huesca
- Intercellular Communication in the Inflammatory Response, Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Cristina Gutiérrez-Vázquez
- Instituto Investigación Sanitaria Princesa (IIS-IP), Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Intercellular Communication in the Inflammatory Response, Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Universidad Autónoma de Madrid, Madrid, Spain
| | - Fátima Sánchez-Cabo
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Jesús Vázquez
- Centro de Investigación Biomédica en Red, Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Štěpánka Vaňáčová
- CEITEC-Central European Institute of Technology, Masaryk University, Kamenice 5/A35, 625 00, Brno, Czech Republic
| | - Francisco Sánchez-Madrid
- Instituto Investigación Sanitaria Princesa (IIS-IP), Immunology Service, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Madrid, Spain.
- Intercellular Communication in the Inflammatory Response, Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
- Universidad Autónoma de Madrid, Madrid, Spain.
- Centro de Investigación Biomédica en Red, Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
| |
Collapse
|
12
|
Law JC, Watts TH. Considerations for Choosing T Cell Assays during a Pandemic. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:169-174. [PMID: 37399079 DOI: 10.4049/jimmunol.2300129] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/15/2023] [Indexed: 07/05/2023]
Abstract
The appropriate immunosurveillance tools are foundational for the creation of therapeutics, vaccines, and containment strategies when faced with outbreaks of novel pathogens. During the COVID-19 pandemic, there was an urgent need to rapidly assess immune memory following infection or vaccination. Although there have been attempts to standardize cellular assays more broadly, methods for measuring cell-mediated immunity remain variable across studies. Commonly used methods include ELISPOT, intracellular cytokine staining, activation-induced markers, cytokine secretion assays, and peptide-MHC tetramer staining. Although each assay offers unique and complementary information on the T cell response, there are challenges associated with standardizing these assays. The choice of assay can be driven by sample size, the need for high throughput, and the information sought. A combination of approaches may be optimal. This review describes the benefits and limitations of commonly used methods for assessing T cell immunity across SARS-CoV-2 studies.
Collapse
Affiliation(s)
- Jaclyn C Law
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Tania H Watts
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
13
|
Du J, Zhang J, Wang L, Wang X, Zhao Y, Lu J, Fan T, Niu M, Zhang J, Cheng F, Li J, Zhu Q, Zhang D, Pei H, Li G, Liang X, Huang H, Cao X, Liu X, Shao W, Sheng J. Selective oxidative protection leads to tissue topological changes orchestrated by macrophage during ulcerative colitis. Nat Commun 2023; 14:3675. [PMID: 37344477 PMCID: PMC10284839 DOI: 10.1038/s41467-023-39173-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 06/01/2023] [Indexed: 06/23/2023] Open
Abstract
Ulcerative colitis is a chronic inflammatory bowel disorder with cellular heterogeneity. To understand the composition and spatial changes of the ulcerative colitis ecosystem, here we use imaging mass cytometry and single-cell RNA sequencing to depict the single-cell landscape of the human colon ecosystem. We find tissue topological changes featured with macrophage disappearance reaction in the ulcerative colitis region, occurring only for tissue-resident macrophages. Reactive oxygen species levels are higher in the ulcerative colitis region, but reactive oxygen species scavenging enzyme SOD2 is barely detected in resident macrophages, resulting in distinct reactive oxygen species vulnerability for inflammatory macrophages and resident macrophages. Inflammatory macrophages replace resident macrophages and cause a spatial shift of TNF production during ulcerative colitis via a cytokine production network formed with T and B cells. Our study suggests components of a mechanism for the observed macrophage disappearance reaction of resident macrophages, providing mechanistic hints for macrophage disappearance reaction in other inflammation or infection situations.
Collapse
Affiliation(s)
- Juan Du
- Department of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China.
| | - Junlei Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
- Zhejiang University Cancer Centre, Zhejiang University, Hangzhou, 310002, China
| | - Lin Wang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
- Zhejiang University Cancer Centre, Zhejiang University, Hangzhou, 310002, China
| | - Xun Wang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
- Zhejiang University Cancer Centre, Zhejiang University, Hangzhou, 310002, China
| | - Yaxing Zhao
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
- Zhejiang University Cancer Centre, Zhejiang University, Hangzhou, 310002, China
| | - Jiaoying Lu
- Department of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
| | - Tingmin Fan
- Department of Clinical Pharmacy, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
- Central Laboratory, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310002, China
| | - Meng Niu
- Department of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
| | - Jie Zhang
- Department of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
| | - Fei Cheng
- Pathology Department, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
| | - Jun Li
- Pathology Department, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
| | - Qi Zhu
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, No. 29 JiangJun Road, Jiang Ning District, Nanjing, Jiangsu, 211106, China
| | - Daoqiang Zhang
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, No. 29 JiangJun Road, Jiang Ning District, Nanjing, Jiangsu, 211106, China
| | - Hao Pei
- MobiDrop (Zhejiang), No. 455 Heshun Road, Tongxiang, Zhejiang, 314500, China
| | - Guang Li
- Department of Gastroenterology, Beijing Chaoyang Hospital, Capital Medical University, Chaoyang District, Beijing, 100024, China
| | - Xingguang Liang
- Department of Clinical Pharmacy, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China
- Central Laboratory, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310002, China
| | - He Huang
- Frontiers Science Center for Synthetic Biology, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, China
| | - Xiaocang Cao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Department of Hepato-Gastroenterology, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, 300000, China.
| | - Xinjuan Liu
- Department of Gastroenterology, Beijing Chaoyang Hospital, Capital Medical University, Chaoyang District, Beijing, 100024, China.
| | - Wei Shao
- College of Computer Science and Technology, Nanjing University of Aeronautics and Astronautics, No. 29 JiangJun Road, Jiang Ning District, Nanjing, Jiangsu, 211106, China.
| | - Jianpeng Sheng
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310002, China.
- Zhejiang University Cancer Centre, Zhejiang University, Hangzhou, 310002, China.
| |
Collapse
|
14
|
Stinchcombe JC, Asano Y, Kaufman CJG, Böhlig K, Peddie CJ, Collinson LM, Nadler A, Griffiths GM. Ectocytosis renders T cell receptor signaling self-limiting at the immune synapse. Science 2023; 380:818-823. [PMID: 37228189 DOI: 10.1126/science.abp8933] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/28/2023] [Indexed: 05/27/2023]
Abstract
Cytotoxic T lymphocytes (CTLs) kill virus-infected and cancer cells through T cell receptor (TCR) recognition. How CTLs terminate signaling and disengage to allow serial killing has remained a mystery. TCR activation triggers membrane specialization within the immune synapse, including the production of diacylglycerol (DAG), a lipid that can induce negative membrane curvature. We found that activated TCRs were shed into DAG-enriched ectosomes at the immune synapse rather than internalized through endocytosis, suggesting that DAG may contribute to the outward budding required for ectocytosis. Budding ectosomes were endocytosed directly by target cells, thereby terminating TCR signaling and simultaneously disengaging the CTL from the target cell to allow serial killing. Thus, ectocytosis renders TCR signaling self-limiting.
Collapse
Affiliation(s)
- Jane C Stinchcombe
- Cambridge Institute for Medical Research, Keith Peters Building, Cambridge CB2 0XY, UK
| | - Yukako Asano
- Cambridge Institute for Medical Research, Keith Peters Building, Cambridge CB2 0XY, UK
| | | | - Kristin Böhlig
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | | | | | - André Nadler
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Gillian M Griffiths
- Cambridge Institute for Medical Research, Keith Peters Building, Cambridge CB2 0XY, UK
| |
Collapse
|
15
|
Park JS, Kim JH, Soh WC, Kim NY, Lee KS, Kim CH, Chung IJ, Lee S, Kim HR, Jun CD. Trogocytic molting of T cell microvilli upregulates T cell receptor surface expression and promotes clonal expansion. Nat Commun 2023; 14:2980. [PMID: 37221214 DOI: 10.1038/s41467-023-38707-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 05/09/2023] [Indexed: 05/25/2023] Open
Abstract
Although T cell activation is known to involve the internalization of the T cell antigen receptor (TCR), much less is known regarding the release of TCRs following T cell interaction with cognate antigen-presenting cells. In this study, we examine the physiological mechanisms underlying TCR release following T cell activation. We show that T cell activation results in the shedding of TCRs in T cell microvilli, which involves a combined process of trogocytosis and enzymatic vesiculation, leading to the loss of membrane TCRs and microvilli-associated proteins and lipids. Surprisingly, unlike TCR internalization, this event results in the rapid upregulation of surface TCR expression and metabolic reprogramming of cholesterol and fatty acid synthesis to support cell division and survival. These results demonstrate that TCRs are lost through trogocytic 'molting' following T cell activation and highlight this mechanism as an important regulator of clonal expansion.
Collapse
Affiliation(s)
- Jeong-Su Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Jun-Hyeong Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Won-Chang Soh
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Na-Young Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Kyung-Sik Lee
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Chang-Hyun Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Ik-Joo Chung
- Department of Hematology-Oncology, Immunotherapy Innovation Center, Chonnam National University Medical School, Hwasun, 58128, Republic of Korea
| | - Sunjae Lee
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Hye-Ran Kim
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea.
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea.
- Division of Rare and Refractory Cancer, Tumor Immunology, Research Institute, National Cancer Center, Goyang, 10408, Republic of Korea.
| | - Chang-Duk Jun
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea.
- Immune Synapse and Cell Therapy Research Center, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea.
| |
Collapse
|
16
|
Charvet B, Brunel J, Pierquin J, Iampietro M, Decimo D, Queruel N, Lucas A, Encabo-Berzosa MDM, Arenaz I, Marmolejo TP, Gonzalez AI, Maldonado AC, Mathieu C, Küry P, Flores-Rivera J, Torres-Ruiz F, Avila-Rios S, Salgado Montes de Oca G, Schoorlemmer J, Perron H, Horvat B. SARS-CoV-2 awakens ancient retroviral genes and the expression of proinflammatory HERV-W envelope protein in COVID-19 patients. iScience 2023; 26:106604. [PMID: 37091988 PMCID: PMC10079620 DOI: 10.1016/j.isci.2023.106604] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 01/29/2023] [Accepted: 03/30/2023] [Indexed: 04/25/2023] Open
Abstract
Patients with COVID-19 may develop abnormal inflammatory response, followed in some cases by severe disease and long-lasting syndromes. We show here that in vitro exposure to SARS-CoV-2 activates the expression of the human endogenous retrovirus (HERV) HERV-W proinflammatory envelope protein (ENV) in peripheral blood mononuclear cells from a subset of healthy donors, in ACE2 receptor and infection-independent manner. Plasma and/or sera of 221 COVID-19 patients from different cohorts, infected with successive SARS-CoV-2 variants including the Omicron, had detectable HERV-W ENV, which correlated with ENV expression in T lymphocytes and peaked with the disease severity. HERV-W ENV was also found in postmortem tissues of lungs, heart, gastrointestinal tract, brain olfactory bulb, and nasal mucosa from COVID-19 patients. Altogether, these results demonstrate that SARS-CoV-2 could induce HERV-W envelope protein expression and suggest its involvement in the immunopathogenesis of certain COVID-19-associated syndromes and thereby its relevance in the development of personalized treatment of patients.
Collapse
Affiliation(s)
| | | | | | - Mathieu Iampietro
- CIRI, International Center for Infectiology Research, INSERM U1111, CNRS UMR5308, Université de Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Didier Decimo
- CIRI, International Center for Infectiology Research, INSERM U1111, CNRS UMR5308, Université de Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | | | - Alexandre Lucas
- We-Met platform, I2MC/Inserm/Université Paul Sabatier UMR1297, Toulouse, France
| | | | - Izaskun Arenaz
- Biobanco del Sistema de Salud de Aragón, Instituto Aragonés de Ciencias de la Salud (IACS), Zaragoza, Spain
| | - Tania Perez Marmolejo
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, México Ciudad, México
| | - Arturo Ivan Gonzalez
- Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, México Ciudad, México
| | | | - Cyrille Mathieu
- CIRI, International Center for Infectiology Research, INSERM U1111, CNRS UMR5308, Université de Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Dusseldorf, Germany
| | - Jose Flores-Rivera
- Department of Neurology, National Institute of Neurology and Neurosurgery, Mexico City, Mexico
| | - Fernanda Torres-Ruiz
- Centro de investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, México Ciudad, México
| | - Santiago Avila-Rios
- Centro de investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, México Ciudad, México
| | - Gonzalo Salgado Montes de Oca
- Centro de investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, México Ciudad, México
| | - Jon Schoorlemmer
- ARAID Fundación; Instituto Aragonés de Ciencias de la Salud (IACS); Grupo B46_20R de la DGA and GIIS-028 del IISA; all Zaragoza, Spain
| | - Hervé Perron
- GeNeuro Innovation, Lyon, France
- GeNeuro, Plan les Ouates, Geneva, Switzerland
| | - Branka Horvat
- CIRI, International Center for Infectiology Research, INSERM U1111, CNRS UMR5308, Université de Lyon, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| |
Collapse
|
17
|
Pura JA, Li X, Chan C, Xie J. TEAM: A MULTIPLE TESTING ALGORITHM ON THE AGGREGATION TREE FOR FLOW CYTOMETRY ANALYSIS. Ann Appl Stat 2023; 17:621-640. [PMID: 38736649 PMCID: PMC11083434 DOI: 10.1214/22-aoas1645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
In immunology studies, flow cytometry is a commonly used multivariate single-cell assay. One key goal in flow cytometry analysis is to detect the immune cells responsive to certain stimuli. Statistically, this problem can be translated into comparing two protein expression probability density functions (pdfs) before and after the stimulus; the goal is to pinpoint the regions where these two pdfs differ. Further screening of these differential regions can be performed to identify enriched sets of responsive cells. In this paper, we model identifying differential density regions as a multiple testing problem. First, we partition the sample space into small bins. In each bin, we form a hypothesis to test the existence of differential pdfs. Second, we develop a novel multiple testing method, called TEAM (Testing on the Aggregation tree Method), to identify those bins that harbor differential pdfs while controlling the false discovery rate (FDR) under the desired level. TEAM embeds the testing procedure into an aggregation tree to test from fine- to coarse-resolution. The procedure achieves the statistical goal of pinpointing density differences to the smallest possible regions. TEAM is computationally efficient, capable of analyzing large flow cytometry data sets in much shorter time compared with competing methods. We applied TEAM and competing methods on a flow cytometry data set to identify T cells responsive to the cytomegalovirus (CMV)-pp65 antigen stimulation. With additional downstream screening, TEAM successfully identified enriched sets containing monofunctional, bifunctional, and polyfunctional T cells. Competing methods either did not finish in a reasonable time frame or provided less interpretable results. Numerical simulations and theoretical justifications demonstrate that TEAM has asymptotically valid, powerful, and robust performance. Overall, TEAM is a computationally efficient and statistically powerful algorithm that can yield meaningful biological insights in flow cytometry studies.
Collapse
Affiliation(s)
- John A Pura
- Center of Innovation to Accelerate Discovery and Practice Transformation, Durham Veterans AfFaIRS MediCal Center, Durham, NC 27701
| | - Xuechan Li
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC 27705
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC 27705
| | - Jichun Xie
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC 27705
| |
Collapse
|
18
|
Rollins MR, Raynor JF, Miller EA, Butler JZ, Spartz EJ, Lahr WS, You Y, Burrack AL, Moriarity BS, Webber BR, Stromnes IM. Germline T cell receptor exchange results in physiological T cell development and function. Nat Commun 2023; 14:528. [PMID: 36726009 PMCID: PMC9892040 DOI: 10.1038/s41467-023-36180-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 01/18/2023] [Indexed: 02/03/2023] Open
Abstract
T cell receptor (TCR) transgenic mice represent an invaluable tool to study antigen-specific immune responses. In the pre-existing models, a monoclonal TCR is driven by a non-physiologic promoter and randomly integrated into the genome. Here, we create a highly efficient methodology to develop T cell receptor exchange (TRex) mice, in which TCRs, specific to the self/tumor antigen mesothelin (Msln), are integrated into the Trac locus, with concomitant Msln disruption to circumvent T cell tolerance. We show that high affinity TRex thymocytes undergo all sequential stages of maturation, express the exogenous TCR at DN4, require MHC class I for positive selection and undergo negative selection only when both Msln alleles are present. By comparison of TCRs with the same specificity but varying affinity, we show that Trac targeting improves functional sensitivity of a lower affinity TCR and confers resistance to T cell functional loss. By generating P14 TRex mice with the same specificity as the widely used LCMV-P14 TCR transgenic mouse, we demonstrate increased avidity of Trac-targeted TCRs over transgenic TCRs, while preserving physiologic T cell development. Together, our results support that the TRex methodology is an advanced tool to study physiological antigen-specific T cell behavior.
Collapse
Affiliation(s)
- Meagan R Rollins
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Jackson F Raynor
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Ebony A Miller
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Jonah Z Butler
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Ellen J Spartz
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
- Department of Medicine, UCLA Health, Los Angeles, CA, USA
| | - Walker S Lahr
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Yun You
- Mouse Genetics Laboratory, University of Minnesota, Minneapolis, MN, USA
| | - Adam L Burrack
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Branden S Moriarity
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Beau R Webber
- Department of Pediatrics, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Ingunn M Stromnes
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA.
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
- Center for Genome Engineering, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
19
|
McCaleb MR, Miranda AM, Ratliff KC, Torres RM, Pelanda R. CD19 Is Internalized Together with IgM in Proportion to B Cell Receptor Stimulation and Is Modulated by Phosphatidylinositol 3-Kinase in Bone Marrow Immature B Cells. Immunohorizons 2023; 7:49-63. [PMID: 36637517 PMCID: PMC10074640 DOI: 10.4049/immunohorizons.2200092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 01/14/2023] Open
Abstract
Newly generated immature B cells that bind self-antigen with high avidity arrest in differentiation and undergo central tolerance via receptor editing and clonal deletion. These autoreactive immature B cells also express low surface levels of the coreceptor CD19, a key activator of the PI3K pathway. Signals emanating from both CD19 and PI3K are known to be critical for attenuating receptor editing and selecting immature B cells into the periphery. However, the mechanisms that modulate CD19 expression at this stage of B cell development have not yet been resolved. Using in vivo and in vitro models, we demonstrate that Cd19 de novo gene transcription and translation do not significantly contribute to the differences in CD19 surface expression in mouse autoreactive and nonautoreactive immature B cells. Instead, CD19 downregulation is induced by BCR stimulation in proportion to BCR engagement, and the remaining surface IgM and CD19 molecules promote intracellular PI3K-AKT activity in proportion to their level of expression. The internalized CD19 is degraded with IgM by the lysosome, but inhibiting lysosome-mediated protein degradation only slightly improves surface CD19. In fact, CD19 is restored only upon Ag removal. Our data also reveal that the PI3K-AKT pathway positively modulates CD19 surface expression in immature B cells via a mechanism that is independent of inhibition of FOXO1 and its role on Cd19 gene transcription while is dependent on mTORC1.
Collapse
Affiliation(s)
- Megan R. McCaleb
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO; and
| | - Anjelica M. Miranda
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO; and
| | - Kaysie C. Ratliff
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO; and
| | - Raul M. Torres
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO; and
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO
| | - Roberta Pelanda
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO; and
- Department of Immunology and Genomic Medicine, National Jewish Health, Denver, CO
| |
Collapse
|
20
|
Cassioli C, Patrussi L, Valitutti S, Baldari CT. Learning from TCR Signaling and Immunological Synapse Assembly to Build New Chimeric Antigen Receptors (CARs). Int J Mol Sci 2022; 23:14255. [PMID: 36430728 PMCID: PMC9694822 DOI: 10.3390/ijms232214255] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/10/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell immunotherapy is a revolutionary pillar in cancer treatment. Clinical experience has shown remarkable successes in the treatment of certain hematological malignancies but only limited efficacy against B cell chronic lymphocytic leukemia (CLL) and other cancer types, especially solid tumors. A wide range of engineering strategies have been employed to overcome the limitations of CAR T cell therapy. However, it has become increasingly clear that CARs have unique, unexpected features; hence, a deep understanding of how CARs signal and trigger the formation of a non-conventional immunological synapse (IS), the signaling platform required for T cell activation and execution of effector functions, would lead a shift from empirical testing to the rational design of new CAR constructs. Here, we review current knowledge of CARs, focusing on their structure, signaling and role in CAR T cell IS assembly. We, moreover, discuss the molecular features accounting for poor responses in CLL patients treated with anti-CD19 CAR T cells and propose CLL as a paradigm for diseases connected to IS dysfunctions that could significantly benefit from the development of novel CARs to generate a productive anti-tumor response.
Collapse
Affiliation(s)
- Chiara Cassioli
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Laura Patrussi
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Salvatore Valitutti
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1037, Centre de Recherche en Cancérologie de Toulouse (CRCT), Université de Toulouse III-Paul Sabatier, 31037 Toulouse, France
- Department of Pathology, Institut Universitaire du Cancer-Oncopole de Toulouse, 31059 Toulouse, France
| | - Cosima T. Baldari
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| |
Collapse
|
21
|
Assessment of T Cell Receptor Complex Expression Kinetics in Natural Killer Cells. Curr Issues Mol Biol 2022; 44:3859-3871. [PMID: 36135177 PMCID: PMC9497757 DOI: 10.3390/cimb44090265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/15/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Among the polypeptides that comprise the T cell receptor (TCR), only CD3ζ is found in Natural Killer (NK) cells, where it transmits signals from activating receptors such as CD16 and NKp46. NK cells are potent immune cells that recognize target cells through germline-encoded activating and inhibitory receptors. Genetic engineering of NK cells enables tumor-specific antigen recognition and, thus, has a significant promise in adoptive cell therapy. Ectopic expression of engineered TCR components in T cells leads to mispairing with the endogenous components, making a knockout of the endogenous TCR necessary. To circumvent the mispairing of TCRs or the need for knockout technologies, TCR complex expression has been studied in NK cells. In the current study, we explored the cellular processing of the TCR complex in NK cells. We observed that in the absence of CD3 subunits, the TCR was not expressed on the surface of NK cells and vice versa. Moreover, a progressive increase in surface expression of TCR between day three and day seven was observed after transduction. Interestingly, the TCR complex expression in NK92 cells was enhanced with a proteasome inhibitor (bortezomib) but not a lysosomal inhibitor (chloroquine). Additionally, we observed that the TCR complex was functional in NK92 cells as measured by estimating CD107a as a degranulation marker, IFNγ cytokine production, and killing assays. NK92 cells strongly degranulated when CD3ε was engaged in the presence of TCR, but not when only CD3 was overexpressed. Therefore, our findings encourage further investigation to unravel the mechanisms that prevent the surface expression of the TCR complex.
Collapse
|
22
|
Purcarea A, Jarosch S, Barton J, Grassmann S, Pachmayr L, D'Ippolito E, Hammel M, Hochholzer A, Wagner KI, van den Berg JH, Buchholz VR, Haanen JBAG, Busch DH, Schober K. Signatures of recent activation identify a circulating T cell compartment containing tumor-specific antigen receptors with high avidity. Sci Immunol 2022; 7:eabm2077. [PMID: 35960818 DOI: 10.1126/sciimmunol.abm2077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
T cell receptor (TCR) avidity is assumed to be a major determinant of the spatiotemporal fate and protective capacity of tumor-specific T cells. However, monitoring polyclonal T cell responses with known TCR avidities in vivo over space and time remains challenging. Here, we investigated the fate and functionality of tumor neoantigen-specific T cells with TCRs of distinct avidities in a well-established, reductionist preclinical tumor model and human patients with melanoma. To this end, we used polyclonal T cell transfers with in-depth characterized TCRs together with flow cytometric phenotyping in mice inoculated with MC38 OVA tumors. Transfer of T cells from retrogenic mice harboring TCRs with high avidity resulted in best tumor protection. Unexpectedly, we found that both high- and low-avidity T cells are similarly abundant within the tumor and adopt concordant phenotypic signs of exhaustion. Outside the tumor, high-avidity TCR T cells were not generally overrepresented but, instead, selectively enriched in T cell populations with intermediate PD-1 protein expression. Single-cell sequencing of neoantigen-specific T cells from two patients with melanoma-combined with transgenic reexpression of identified TCRs by CRISPR-Cas9-mediated orthotopic TCR replacement-revealed high-functionality TCRs to be enriched in T cells with RNA signatures of recent activation. Furthermore, of 130 surface protein candidates, PD-1 surface expression was most consistently enriched in functional TCRs. Together, our findings show that tumor-reactive TCRs with high protective capacity circulating in peripheral blood are characterized by a signature of recent activation.
Collapse
Affiliation(s)
- Anna Purcarea
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Sebastian Jarosch
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Jack Barton
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Simon Grassmann
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Ludwig Pachmayr
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Elvira D'Ippolito
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Monika Hammel
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Anna Hochholzer
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Karolin I Wagner
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany
| | | | - Veit R Buchholz
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - John B A G Haanen
- Division of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Dirk H Busch
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany.,German Center for Infection Research (DZIF), Munich, Germany.,Focus Group "Clinical Cell Processing and Purification", Institute for Advanced Study, TUM, Munich, Germany
| | - Kilian Schober
- Institute for Medical Microbiology, Immunology, and Hygiene, Technische Universität München (TUM), Munich, Germany.,Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie, und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
23
|
Dobson CS, Reich AN, Gaglione S, Smith BE, Kim EJ, Dong J, Ronsard L, Okonkwo V, Lingwood D, Dougan M, Dougan SK, Birnbaum ME. Antigen identification and high-throughput interaction mapping by reprogramming viral entry. Nat Methods 2022; 19:449-460. [PMID: 35396484 PMCID: PMC9012700 DOI: 10.1038/s41592-022-01436-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 03/01/2022] [Indexed: 01/11/2023]
Abstract
Deciphering immune recognition is critical for understanding a broad range of diseases and for the development of effective vaccines and immunotherapies. Efforts to do so are limited by a lack of technologies capable of simultaneously capturing the complexity of adaptive immunoreceptor repertoires and the landscape of potential antigens. To address this, we present receptor-antigen pairing by targeted retroviruses, which combines viral pseudotyping and molecular engineering approaches to enable one-pot library-on-library interaction screens by displaying antigens on the surface of lentiviruses and encoding their identity in the viral genome. Antigen-specific viral infection of cell lines expressing human T or B cell receptors allows readout of both antigen and receptor identities via single-cell sequencing. The resulting system is modular, scalable and compatible with any cell type. These techniques provide a suite of tools for targeted viral entry, molecular engineering and interaction screens with broad potential applications.
Collapse
Affiliation(s)
- Connor S Dobson
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | - Anna N Reich
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | - Stephanie Gaglione
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Blake E Smith
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
- Program in Immunology, Harvard Medical School, Boston, MA, USA
| | - Ellen J Kim
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | - Jiayi Dong
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA
| | | | - Vintus Okonkwo
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | | | - Michael Dougan
- Program in Immunology, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Stephanie K Dougan
- Program in Immunology, Harvard Medical School, Boston, MA, USA
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael E Birnbaum
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA.
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
- Singapore-MIT Alliance for Research and Technology Centre, Singapore, Singapore.
| |
Collapse
|
24
|
Jayasooriya V, Ringwelski B, Dorsam G, Nawarathna D. mRNA-based CAR T-cells manufactured by miniaturized two-step electroporation produce selective cytotoxicity toward target cancer cells. LAB ON A CHIP 2021; 21:3748-3761. [PMID: 34585697 PMCID: PMC8513750 DOI: 10.1039/d1lc00219h] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
There is a growing interest for viral vector-free chimeric antigen receptor (CAR) T-cells due to its ability to kill cancer cells without adverse side effects. A potential avenue for manufacturing viral-vector free CAR T-cells is to utilize mRNA electroporation. One of the major concerns with mRNA electroporated CAR T-cells is the shorter cytotoxic lifespan of a few days, which is insufficient or not ideal for therapy. To better understand this issue and develop a potential solution, this study focused on examining the translation of electroporated mRNA to CAR molecules, time dependent degradation of CAR molecules and cytotoxicity produced by CAR T-cells on cancer cells. It was found that the initial expression of CAR molecules dictates the cytotoxicity. Initial CAR expression could be controlled by the experimental parameters such as electroporation time and mRNA concentration in the electroporation buffer. Experiments were carried out using a novel two-step electroporation that allows for controlled and uniform transfection of T-cells. These technical advancements and subsequent findings could provide a viable path for producing CAR T-cells with longer cytotoxic lifespans.
Collapse
Affiliation(s)
- Vidura Jayasooriya
- Department of Electrical and Computer Engineering, North Dakota State University, Fargo, North Dakota, USA.
| | - Beth Ringwelski
- Biomedical Engineering Program, North Dakota State University, Fargo, North Dakota, USA
| | - Glenn Dorsam
- Department of Microbiological Sciences, North Dakota State University, Fargo, ND, 58102, USA
| | - Dharmakeerthi Nawarathna
- Department of Electrical and Computer Engineering, North Dakota State University, Fargo, North Dakota, USA.
- Biomedical Engineering Program, North Dakota State University, Fargo, North Dakota, USA
| |
Collapse
|
25
|
AuYeung AWK, Mould RC, Stegelmeier AA, van Vloten JP, Karimi K, Woods JP, Petrik JJ, Wood GA, Bridle BW. Mechanisms that allow vaccination against an oncolytic vesicular stomatitis virus-encoded transgene to enhance safety without abrogating oncolysis. Sci Rep 2021; 11:15290. [PMID: 34315959 PMCID: PMC8316323 DOI: 10.1038/s41598-021-94483-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/09/2021] [Indexed: 11/26/2022] Open
Abstract
Vaccination can prevent viral infections via virus-specific T cells, among other mechanisms. A goal of oncolytic virotherapy is replication of oncolytic viruses (OVs) in tumors, so pre-existing T cell immunity against an OV-encoded transgene would seem counterproductive. We developed a treatment for melanomas by pre-vaccinating against an oncolytic vesicular stomatitis virus (VSV)-encoded tumor antigen. Surprisingly, when the VSV-vectored booster vaccine was administered at the peak of the primary effector T cell response, oncolysis was not abrogated. We sought to determine how oncolysis was retained during a robust T cell response against the VSV-encoded transgene product. A murine melanoma model was used to identify two mechanisms that enable this phenomenon. First, tumor-infiltrating T cells had reduced cytopathic potential due to immunosuppression. Second, virus-induced lymphopenia acutely removed virus-specific T cells from tumors. These mechanisms provide a window of opportunity for replication of oncolytic VSV and rationale for a paradigm change in oncolytic virotherapy, whereby immune responses could be intentionally induced against a VSV-encoded melanoma-associated antigen to improve safety without abrogating oncolysis.
Collapse
Affiliation(s)
- Amanda W K AuYeung
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Robert C Mould
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Ashley A Stegelmeier
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Jacob P van Vloten
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Khalil Karimi
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - J Paul Woods
- Department of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - James J Petrik
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Geoffrey A Wood
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Byram W Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada. .,Department of Pathobiology, Ontario Veterinary College, University of Guelph, Rm. 4834, Bldg. 89, 50 Stone Rd. E., Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
26
|
van der Donk LEH, Ates LS, van der Spek J, Tukker LM, Geijtenbeek TBH, van Heijst JWJ. Separate signaling events control TCR downregulation and T cell activation in primary human T cells. IMMUNITY INFLAMMATION AND DISEASE 2020; 9:223-238. [PMID: 33350598 PMCID: PMC7860602 DOI: 10.1002/iid3.383] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/27/2020] [Accepted: 11/17/2020] [Indexed: 12/02/2022]
Abstract
Introduction T‐cell antigen receptor (TCR) interaction with cognate peptide:MHC complexes trigger clustering of TCR:CD3 complexes and signal transduction. Triggered TCR:CD3 complexes are rapidly internalized and degraded in a process called ligand‐induced TCR downregulation. Classic studies in immortalized T‐cell lines have revealed a major role for the Src family kinase Lck in TCR downregulation. However, to what extent a similar mechanism operates in primary human T cells remains unclear. Methods Here, we developed an anti‐CD3‐mediated TCR downregulation assay, in which T‐cell gene expression in primary human T cells can be knocked down by microRNA constructs. In parallel, we used CRISPR/Cas9‐mediated knockout in Jurkat cells for validation experiments. Results We efficiently knocked down the expression of tyrosine kinases Lck, Fyn, and ZAP70, and found that, whereas this impaired T cell activation and effector function, TCR downregulation was not affected. Although TCR downregulation was marginally inhibited by the simultaneous knockdown of Lck and Fyn, its full abrogation required broad‐acting tyrosine kinase inhibitors. Conclusions These data suggest that there is substantial redundancy in the contribution of individual tyrosine kinases to TCR downregulation in primary human T cells. Our results highlight that TCR downregulation and T cell activation are controlled by different signaling events and illustrate the need for further research to untangle these processes.
Collapse
Affiliation(s)
- Lieve E H van der Donk
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Louis S Ates
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jet van der Spek
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Laura M Tukker
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Teunis B H Geijtenbeek
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Jeroen W J van Heijst
- Department of Experimental Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Neogene Therapeutics, Amsterdam, The Netherlands
| |
Collapse
|
27
|
Baugh R, Khalique H, Seymour LW. Convergent Evolution by Cancer and Viruses in Evading the NKG2D Immune Response. Cancers (Basel) 2020; 12:E3827. [PMID: 33352921 PMCID: PMC7766243 DOI: 10.3390/cancers12123827] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/11/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
The natural killer group 2 member D (NKG2D) receptor and its family of NKG2D ligands (NKG2DLs) are key components in the innate immune system, triggering NK, γδ and CD8+ T cell-mediated immune responses. While surface NKG2DL are rarely found on healthy cells, expression is significantly increased in response to various types of cellular stress, viral infection, and tumour cell transformation. In order to evade immune-mediated cytotoxicity, both pathogenic viruses and cancer cells have evolved various mechanisms of subverting immune defences and preventing NKG2DL expression. Comparisons of the mechanisms employed following virus infection or malignant transformation reveal a pattern of converging evolution at many of the key regulatory steps involved in NKG2DL expression and subsequent immune responses. Exploring ways to target these shared steps in virus- and cancer-mediated immune evasion may provide new mechanistic insights and therapeutic opportunities, for example, using oncolytic virotherapy to re-engage the innate immune system towards cancer cells.
Collapse
Affiliation(s)
| | | | - Leonard W. Seymour
- Anticancer Viruses and Cancer Vaccines Research Group, Department of Oncology, University of Oxford, Oxford OX3 7DQ, UK; (R.B.); (H.K.)
| |
Collapse
|
28
|
Mastrogiovanni M, Juzans M, Alcover A, Di Bartolo V. Coordinating Cytoskeleton and Molecular Traffic in T Cell Migration, Activation, and Effector Functions. Front Cell Dev Biol 2020; 8:591348. [PMID: 33195256 PMCID: PMC7609836 DOI: 10.3389/fcell.2020.591348] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/24/2020] [Indexed: 12/28/2022] Open
Abstract
Dynamic localization of receptors and signaling molecules at the plasma membrane and within intracellular vesicular compartments is crucial for T lymphocyte sensing environmental cues, triggering membrane receptors, recruiting signaling molecules, and fine-tuning of intracellular signals. The orchestrated action of actin and microtubule cytoskeleton and intracellular vesicle traffic plays a key role in all these events that together ensure important steps in T cell physiology. These include extravasation and migration through lymphoid and peripheral tissues, T cell interactions with antigen-presenting cells, T cell receptor (TCR) triggering by cognate antigen-major histocompatibility complex (MHC) complexes, immunological synapse formation, cell activation, and effector functions. Cytoskeletal and vesicle traffic dynamics and their interplay are coordinated by a variety of regulatory molecules. Among them, polarity regulators and membrane-cytoskeleton linkers are master controllers of this interplay. Here, we review the various ways the T cell plasma membrane, receptors, and their signaling machinery interplay with the actin and microtubule cytoskeleton and with intracellular vesicular compartments. We highlight the importance of this fine-tuned crosstalk in three key stages of T cell biology involving cell polarization: T cell migration in response to chemokines, immunological synapse formation in response to antigen cues, and effector functions. Finally, we discuss two examples of perturbation of this interplay in pathological settings, such as HIV-1 infection and mutation of the polarity regulator and tumor suppressor adenomatous polyposis coli (Apc) that leads to familial polyposis and colorectal cancer.
Collapse
Affiliation(s)
- Marta Mastrogiovanni
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
- Collège Doctoral, Sorbonne Université, Paris, France
| | - Marie Juzans
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Andrés Alcover
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
| | - Vincenzo Di Bartolo
- Ligue Nationale Contre le Cancer – Equipe Labellisée LIGUE 2018, Lymphocyte Cell Biology Unit, INSERM-U1221, Department of Immunology, Institut Pasteur, Paris, France
| |
Collapse
|
29
|
Grzywa TM, Sosnowska A, Matryba P, Rydzynska Z, Jasinski M, Nowis D, Golab J. Myeloid Cell-Derived Arginase in Cancer Immune Response. Front Immunol 2020; 11:938. [PMID: 32499785 PMCID: PMC7242730 DOI: 10.3389/fimmu.2020.00938] [Citation(s) in RCA: 273] [Impact Index Per Article: 54.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 04/22/2020] [Indexed: 12/13/2022] Open
Abstract
Amino acid metabolism is a critical regulator of the immune response, and its modulating becomes a promising approach in various forms of immunotherapy. Insufficient concentrations of essential amino acids restrict T-cells activation and proliferation. However, only arginases, that degrade L-arginine, as well as enzymes that hydrolyze L-tryptophan are substantially increased in cancer. Two arginase isoforms, ARG1 and ARG2, have been found to be present in tumors and their increased activity usually correlates with more advanced disease and worse clinical prognosis. Nearly all types of myeloid cells were reported to produce arginases and the increased numbers of various populations of myeloid-derived suppressor cells and macrophages correlate with inferior clinical outcomes of cancer patients. Here, we describe the role of arginases produced by myeloid cells in regulating various populations of immune cells, discuss molecular mechanisms of immunoregulatory processes involving L-arginine metabolism and outline therapeutic approaches to mitigate the negative effects of arginases on antitumor immune response. Development of potent arginase inhibitors, with improved pharmacokinetic properties, may lead to the elaboration of novel therapeutic strategies based on targeting immunoregulatory pathways controlled by L-arginine degradation.
Collapse
Affiliation(s)
- Tomasz M. Grzywa
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Anna Sosnowska
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Paweł Matryba
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Laboratory of Neurobiology BRAINCITY, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Warsaw, Poland
- The Doctoral School of the Medical University of Warsaw, Medical University of Warsaw, Warsaw, Poland
| | - Zuzanna Rydzynska
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Marcin Jasinski
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Dominika Nowis
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Laboratory of Experimental Medicine, Center of New Technologies, University of Warsaw, Warsaw, Poland
- Genomic Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Jakub Golab
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Centre of Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
30
|
Pedrera M, Macchi F, McLean RK, Franceschi V, Thakur N, Russo L, Medfai L, Todd S, Tchilian EZ, Audonnet JC, Chappell K, Isaacs A, Watterson D, Young PR, Marsh GA, Bailey D, Graham SP, Donofrio G. Bovine Herpesvirus-4-Vectored Delivery of Nipah Virus Glycoproteins Enhances T Cell Immunogenicity in Pigs. Vaccines (Basel) 2020; 8:vaccines8010115. [PMID: 32131403 PMCID: PMC7157636 DOI: 10.3390/vaccines8010115] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 02/24/2020] [Accepted: 02/27/2020] [Indexed: 02/07/2023] Open
Abstract
Nipah virus (NiV) is an emergent pathogen capable of causing acute respiratory illness and fatal encephalitis in pigs and humans. A high fatality rate and broad host tropism makes NiV a serious public and animal health concern. There is therefore an urgent need for a NiV vaccines to protect animals and humans. In this study we investigated the immunogenicity of bovine herpesvirus (BoHV-4) vectors expressing either NiV attachment (G) or fusion (F) glycoproteins, BoHV-4-A-CMV-NiV-GΔTK or BoHV-4-A-CMV-NiV-FΔTK, respectively in pigs. The vaccines were benchmarked against a canarypox (ALVAC) vector expressing NiV G, previously demonstrated to induce protective immunity in pigs. Both BoHV-4 vectors induced robust antigen-specific antibody responses. BoHV-4-A-CMV-NiV-GΔTK stimulated NiV-neutralizing antibody titers comparable to ALVAC NiV G and greater than those induced by BoHV-4-A-CMV-NiV-FΔTK. In contrast, only BoHV-4-A-CMV-NiV-FΔTK immunized pigs had antibodies capable of significantly neutralizing NiV G and F-mediated cell fusion. All three vectored vaccines evoked antigen-specific CD4 and CD8 T cell responses, which were particularly strong in BoHV-4-A-CMV-NiV-GΔTK immunized pigs and to a lesser extent BoHV-4-A-CMV-NiV-FΔTK. These findings emphasize the potential of BoHV-4 vectors for inducing antibody and cell-mediated immunity in pigs and provide a solid basis for the further evaluation of these vectored NiV vaccine candidates.
Collapse
Affiliation(s)
- Miriam Pedrera
- The Pirbright Institute, Ash Road, Pirbright GU24 0NF, UK; (M.P.); (R.K.M.); (N.T.); (L.M.); (E.Z.T.); (D.B.)
| | - Francesca Macchi
- Department of Medical-Veterinary Science, University of Parma, 43126 Parma, Italy; (F.M.); (V.F.); (L.R.)
| | - Rebecca K. McLean
- The Pirbright Institute, Ash Road, Pirbright GU24 0NF, UK; (M.P.); (R.K.M.); (N.T.); (L.M.); (E.Z.T.); (D.B.)
| | - Valentina Franceschi
- Department of Medical-Veterinary Science, University of Parma, 43126 Parma, Italy; (F.M.); (V.F.); (L.R.)
| | - Nazia Thakur
- The Pirbright Institute, Ash Road, Pirbright GU24 0NF, UK; (M.P.); (R.K.M.); (N.T.); (L.M.); (E.Z.T.); (D.B.)
| | - Luca Russo
- Department of Medical-Veterinary Science, University of Parma, 43126 Parma, Italy; (F.M.); (V.F.); (L.R.)
| | - Lobna Medfai
- The Pirbright Institute, Ash Road, Pirbright GU24 0NF, UK; (M.P.); (R.K.M.); (N.T.); (L.M.); (E.Z.T.); (D.B.)
- UnivLyon, Université Claude Bernard Lyon 1, 69100 Villeurbanne, France
| | - Shawn Todd
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria 3219, Australia; (S.T.); (G.A.M.)
| | - Elma Z. Tchilian
- The Pirbright Institute, Ash Road, Pirbright GU24 0NF, UK; (M.P.); (R.K.M.); (N.T.); (L.M.); (E.Z.T.); (D.B.)
| | - Jean-Christophe Audonnet
- Boehringer Ingelheim Animal Health, Bâtiment 700 R&D, 813 Cours du 3ème Millénaire, 69800 Saint Priest, France;
| | - Keith Chappell
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Queensland 4072, Australia; (K.C.); (A.I.); (D.W.); (P.R.Y.)
| | - Ariel Isaacs
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Queensland 4072, Australia; (K.C.); (A.I.); (D.W.); (P.R.Y.)
| | - Daniel Watterson
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Queensland 4072, Australia; (K.C.); (A.I.); (D.W.); (P.R.Y.)
| | - Paul R. Young
- Australian Infectious Diseases Research Centre, School of Chemistry and Molecular Biosciences, University of Queensland, St. Lucia, Queensland 4072, Australia; (K.C.); (A.I.); (D.W.); (P.R.Y.)
| | - Glenn A. Marsh
- CSIRO Health and Biosecurity, Australian Animal Health Laboratory, Geelong, Victoria 3219, Australia; (S.T.); (G.A.M.)
| | - Dalan Bailey
- The Pirbright Institute, Ash Road, Pirbright GU24 0NF, UK; (M.P.); (R.K.M.); (N.T.); (L.M.); (E.Z.T.); (D.B.)
| | - Simon P. Graham
- The Pirbright Institute, Ash Road, Pirbright GU24 0NF, UK; (M.P.); (R.K.M.); (N.T.); (L.M.); (E.Z.T.); (D.B.)
- Correspondence: (S.P.G.); (G.D.)
| | - Gaetano Donofrio
- Department of Medical-Veterinary Science, University of Parma, 43126 Parma, Italy; (F.M.); (V.F.); (L.R.)
- Correspondence: (S.P.G.); (G.D.)
| |
Collapse
|
31
|
Yamada S, Miyoshi S, Nishio J, Mizutani S, Yamada Z, Kusunoki N, Sato H, Kuboi Y, Hoshino-Negishi K, Ishii N, Imai T, Mikami T, Nakano H, Kawai S, Nanki T. Effects of CX3CL1 inhibition on murine bleomycin-induced interstitial pneumonia. EUR J INFLAMM 2020. [DOI: 10.1177/2058739220959903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: Treatment for interstitial pneumonia (IP) associated with collagen diseases has not been established. There is a need to elucidate the pathogenesis of IP and develop a novel therapy. We aimed to clarify the role of chemokine (C-X3-C motif) ligand 1 (CX3CL1, also known as fractalkine) in IP. Methods: Bleomycin (BLM) was intratracheally administered to C57BL/6 mice to induce IP. For treatment with control Ab or anti-CX3CL1 mAb, the mice were administered either Ab three times per week for 2 weeks from the day of BLM administration until euthanasia. Expressions of CX3CL1 and its unique receptor CX3CR1 in the lung tissue were examined by immunohistochemical analysis. Cellular infiltration and lung fibrosis were evaluated based on hematoxylin-eosin-staining and Sirius red staining of the lung tissue sections, respectively. Bronchoalveolar lavage fluid (BALF) cells were analyzed by flow cytometry. Results: CX3CL1 and CX3CR1 were strongly expressed in the lung tissue from mice with BLM-induced IP (BLM-IP). Treatment with anti-CX3CL1 mAb did not significantly alter inflammatory cell infiltration or fibrosis in the lung tissue. However, the number of M1-like macrophages in BALF was decreased and surface CD3 expression on T cells was increased by anti-CX3CL1 mAb treatment. Conclusions: Inhibition of CX3CL1 decreased inflammatory cells and may attenuate T cell activation in BALF. CX3CL1 inhibitor may have the potential to suppress the infiltration and activation of immune cells in IP.
Collapse
Affiliation(s)
- Soichi Yamada
- Department of Internal Medicine, Toho University Graduate School of Medicine, Ota-ku, Tokyo, Japan
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, Ota-ku, Tokyo, Japan
| | - Shion Miyoshi
- Department of Respiratory Medicine, Toho University Graduate School of Medicine, Ota-ku, Tokyo, Japan
- Department of Biochemistry, Toho University School of Medicine, Ota-ku, Tokyo, Japan
| | - Junko Nishio
- Department of Immunopathology and Immunoregulation, Toho University School of Medicine, Ota-ku, Tokyo, Japan
| | - Satoshi Mizutani
- Department of Internal Medicine, Toho University Graduate School of Medicine, Ota-ku, Tokyo, Japan
| | - Zento Yamada
- Department of Internal Medicine, Toho University Graduate School of Medicine, Ota-ku, Tokyo, Japan
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, Ota-ku, Tokyo, Japan
| | - Natsuko Kusunoki
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, Ota-ku, Tokyo, Japan
- Department of Inflammation and Pain Control Research, Toho University School of Medicine, Ota-ku, Tokyo, Japan
| | - Hiroshi Sato
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, Ota-ku, Tokyo, Japan
| | | | | | - Naoto Ishii
- KAN Research Institute, Inc., Kobe, Hyogo, Japan
| | - Toshio Imai
- KAN Research Institute, Inc., Kobe, Hyogo, Japan
| | - Tetsuo Mikami
- Department of Pathology, Toho University School of Medicine, Ota-ku, Tokyo, Japan
| | - Hiroyasu Nakano
- Department of Biochemistry, Toho University School of Medicine, Ota-ku, Tokyo, Japan
| | - Shinichi Kawai
- Department of Inflammation and Pain Control Research, Toho University School of Medicine, Ota-ku, Tokyo, Japan
| | - Toshihiro Nanki
- Department of Internal Medicine, Toho University Graduate School of Medicine, Ota-ku, Tokyo, Japan
- Division of Rheumatology, Department of Internal Medicine, Toho University School of Medicine, Ota-ku, Tokyo, Japan
| |
Collapse
|
32
|
Mitsuiki N, Schwab C, Grimbacher B. What did we learn from CTLA-4 insufficiency on the human immune system? Immunol Rev 2019; 287:33-49. [PMID: 30565239 DOI: 10.1111/imr.12721] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Accepted: 09/16/2018] [Indexed: 02/07/2023]
Abstract
Cytotoxic-T-lymphocyte-antigen-4 (CTLA-4) is a negative immune regulator constitutively expressed on regulatory T (Treg) cells and upregulated on activated T cells. CTLA-4 inhibits T cell activation by various suppressive functions including competition with CD28, regulation of the inhibitory function of Treg cells, such as transendocytosis, and the control of adhesion and motility. Intrinsic CTLA-4 signaling has been controversially discussed, but so far no distinct signaling pathway has been identified. The CTLA-4-mediated Treg suppression plays an important role in the maintenance of peripheral tolerance and the prevention of autoimmune diseases. Human CTLA-4 insufficiency is caused by heterozygous germline mutations in CTLA4 and characterized by a complex immune dysregulation syndrome. Clinical studies on CTLA4 mutation carriers showed a reduced penetrance and variable expressivity, suggesting modifying factor(s). One hundred and forty-eight CTLA4 mutation carriers have been reported; patients showed hypogammaglobulinemia, recurrent infectious diseases, various autoimmune diseases, and lymphocytic infiltration into multiple organs. The CTLA-4 expression level in Treg cells was reduced, while the frequency of Treg cells was increased in CTLA-4-insufficient patients. The transendocytosis assay is a specific functional test for the assessment of newly identified CTLA4 gene variants. Immunoglobulin substitution, corticosteroids, immunosuppressive therapy, and targeted therapy such as with CTLA-4 fusion proteins and mechanistic target of rapamycin (mTOR) inhibitors were applied; patients with life-threatening, treatment-resistant symptoms underwent hematopoietic stem cell transplantation. The fact that in humans CTLA-4 insufficiency causes severe disease taught us that the amount of CTLA-4 molecules present in/on T cells matters for immune homeostasis. However, whether the pathology-causing activated T lymphocytes in CTLA-4-insufficient patients are antigen-specific is an unsolved question. CTLA-4, in addition, has a role in autoimmune diseases and cancer. Anti-CTLA-4 drugs are employed as checkpoint inhibitors to target various forms of cancer. Thus, clinical research on human CTLA-4 insufficiency might provide us a deeper understanding of the mechanism(s) of the CTLA-4 molecule and immune dysregulation disorders.
Collapse
Affiliation(s)
- Noriko Mitsuiki
- Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Charlotte Schwab
- Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bodo Grimbacher
- Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
33
|
Duong MN, Erdes E, Hebeisen M, Rufer N. Chronic TCR-MHC (self)-interactions limit the functional potential of TCR affinity-increased CD8 T lymphocytes. J Immunother Cancer 2019; 7:284. [PMID: 31690351 PMCID: PMC6833194 DOI: 10.1186/s40425-019-0773-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 10/05/2019] [Indexed: 12/21/2022] Open
Abstract
Background Affinity-optimized T cell receptor (TCR)-engineered lymphocytes targeting tumor antigens can mediate potent antitumor responses in cancer patients, but also bear substantial risks for off-target toxicities. Most preclinical studies have focused on T cell responses to antigen-specific stimulation. In contrast, little is known on the regulation of T cell responsiveness through continuous TCR triggering and consequent tonic signaling. Here, we addressed the question whether increasing the TCR affinity can lead to chronic interactions occurring directly between TCRs and MHC-(self) molecules, which may modulate the overall functional potency of tumor-redirected CD8 T cells. For this purpose, we developed two complementary human CD8 T cell models (i.e. HLA-A2 knock-in and knock-out) engineered with incremental-affinity TCRs to the HLA-A2/NY-ESO-1 tumor antigen. Methods The impact of HLA-A2 recognition, depending on TCR affinity, was assessed at the levels of the TCR/CD3 complex, regulatory receptors, and signaling, under steady-state conditions and in kinetic studies. The quality of CD8 T cell responses was further evaluated by gene expression and multiplex cytokine profiling, as well as real-time quantitative cell killing, combined with co-culture assays. Results We found that HLA-A2 per se (in absence of cognate peptide) can trigger chronic activation followed by a tolerance-like state of tumor-redirected CD8 T cells with increased-affinity TCRs. HLA-A2pos but not HLA-A2neg T cells displayed an activation phenotype, associated with enhanced upregulation of c-CBL and multiple inhibitory receptors. T cell activation preceded TCR/CD3 downmodulation, impaired TCR signaling and functional hyporesponsiveness. This stepwise activation-to-hyporesponsive state was dependent on TCR affinity and already detectable at the upper end of the physiological affinity range (KD ≤ 1 μM). Similar findings were made when affinity-increased HLA-A2neg CD8 T cells were chronically exposed to HLA-A2pos-expressing target cells. Conclusions Our observations indicate that sustained interactions between affinity-increased TCR and self-MHC can directly adjust the functional potential of T cells, even in the absence of antigen-specific stimulation. The observed tolerance-like state depends on TCR affinity and has therefore potential implications for the design of affinity-improved TCRs for adoptive T cell therapy, as several engineered TCRs currently used in clinical trials share similar affinity properties.
Collapse
Affiliation(s)
- Minh Ngoc Duong
- Department of oncology UNIL CHUV, Lausanne University Hospital and University of Lausanne, CH-1066, Epalinges, Switzerland
| | - Efe Erdes
- Department of oncology UNIL CHUV, Lausanne University Hospital and University of Lausanne, CH-1066, Epalinges, Switzerland
| | - Michael Hebeisen
- Department of oncology UNIL CHUV, Lausanne University Hospital and University of Lausanne, CH-1066, Epalinges, Switzerland.
| | - Nathalie Rufer
- Department of oncology UNIL CHUV, Lausanne University Hospital and University of Lausanne, CH-1066, Epalinges, Switzerland.
| |
Collapse
|
34
|
Rossatti P, Ziegler L, Schregle R, Betzler VM, Ecker M, Rossy J. Cdc42 Couples T Cell Receptor Endocytosis to GRAF1-Mediated Tubular Invaginations of the Plasma Membrane. Cells 2019; 8:cells8111388. [PMID: 31690048 PMCID: PMC6912536 DOI: 10.3390/cells8111388] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 10/31/2019] [Accepted: 10/31/2019] [Indexed: 02/07/2023] Open
Abstract
: T cell activation is immediately followed by internalization of the T cell receptor (TCR). TCR endocytosis is required for T cell activation, but the mechanisms supporting removal of TCR from the cell surface remain incompletely understood. Here we report that TCR endocytosis is linked to the clathrin-independent carrier (CLIC) and GPI-enriched endocytic compartments (GEEC) endocytic pathway. We show that unlike the canonical clathrin cargo transferrin or the adaptor protein Lat, internalized TCR accumulates in tubules shaped by the small GTPase Cdc42 and the Bin/amphiphysin/Rvs (BAR) domain containing protein GRAF1 in T cells. Preventing GRAF1-positive tubules to mature into endocytic vesicles by expressing a constitutively active Cdc42 impairs the endocytosis of TCR, while having no consequence on the uptake of transferrin. Together, our data reveal a link between TCR internalization and the CLIC/GEEC endocytic route supported by Cdc42 and GRAF1.
Collapse
Affiliation(s)
- Pascal Rossatti
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, 8280 Kreuzlingen, Switzerland.
| | - Luca Ziegler
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, 8280 Kreuzlingen, Switzerland.
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany.
| | - Richard Schregle
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, 8280 Kreuzlingen, Switzerland.
| | - Verena M Betzler
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, 8280 Kreuzlingen, Switzerland.
| | - Manuela Ecker
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, University of New South Wales, Sydney 2052, Australia.
| | - Jérémie Rossy
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, 8280 Kreuzlingen, Switzerland.
- Department of Biology, University of Konstanz, 78457 Konstanz, Germany.
| |
Collapse
|
35
|
Cryopreserved Human Natural Killer Cells Exhibit Potent Antitumor Efficacy against Orthotopic Pancreatic Cancer through Efficient Tumor-Homing and Cytolytic Ability (Running Title: Cryopreserved NK Cells Exhibit Antitumor Effect). Cancers (Basel) 2019; 11:cancers11070966. [PMID: 31324057 PMCID: PMC6678894 DOI: 10.3390/cancers11070966] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Revised: 07/04/2019] [Accepted: 07/05/2019] [Indexed: 12/30/2022] Open
Abstract
Pancreatic cancer is known to be highly aggressive, and desmoplasia-induced accumulation of extracellular matrix (ECM), which is a hallmark of many pancreatic cancers, severely restricts the therapeutic efficacy of both immunotherapeutics and conventional chemotherapeutics due to the ECM functioning as a major physical barrier against permeation and penetration. In the case of cell-based immunotherapeutics, there are several other bottlenecks preventing translation into clinical use due to their biological nature; for example, poor availability of cell therapeutic in a readily usable form due to difficulties in production, handling, shipping, and storage. To address these challenges, we have isolated allogeneic natural killer (NK) cells from healthy donors and expanded them in vitro to generate cryopreserved stocks. These cryopreserved NK cells were thawed to evaluate their therapeutic efficacy against desmoplastic pancreatic tumors, ultimately aiming to develop a readily accessible and mass-producible off-the-shelf cell-based immunotherapeutic. The cultured NK cells post-thawing retained highly pure populations of activated NK cells that expressed various activating receptors and a chemokine receptor. Furthermore, systemic administration of NK cells induced greater in vivo tumor growth suppression when compared with gemcitabine, which is the standard chemotherapeutic used for pancreatic cancer treatment. The potent antitumor effect of NK cells was mediated by efficient tumor-homing ability and infiltration into desmoplastic tumor tissues. Moreover, the infiltration of NK cells led to strong induction of apoptosis, elevated expression of the antitumor cytokine interferon (IFN)-γ, and inhibited expression of the immunosuppressive transforming growth factor (TGF)-β in tumor tissues. Expanded and cryopreserved NK cells are strong candidates for future cell-mediated systemic immunotherapy against pancreatic cancer.
Collapse
|
36
|
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
|
37
|
Boesch M, Cosma A, Sopper S. Flow Cytometry: To Dump or Not To Dump. THE JOURNAL OF IMMUNOLOGY 2018; 201:1813-1815. [DOI: 10.4049/jimmunol.1801037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 07/26/2018] [Indexed: 12/20/2022]
|
38
|
Zuo J, Mohammed F, Moss P. The Biological Influence and Clinical Relevance of Polymorphism Within the NKG2D Ligands. Front Immunol 2018; 9:1820. [PMID: 30166984 PMCID: PMC6105697 DOI: 10.3389/fimmu.2018.01820] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 07/24/2018] [Indexed: 01/23/2023] Open
Abstract
NKG2D is a major regulator of the activity of cytotoxic cells and interacts with eight different ligands (NKG2DL) from two families of MIC and ULBP proteins. The selective forces that drove evolution of NKG2DL are uncertain, but are likely to have been dominated by infectious disease and cancer. Of interest, NKG2DL are some of the most polymorphic genes outside the MHC locus and the study of these is uncovering a range of novel observations regarding the structure and function of NKG2DL. Polymorphism is present within all NKG2DL members and varies markedly within different populations. Allelic variation influences functional responses through three major mechanisms. First, it may drive differential levels of protein expression, modulate subcellular trafficking, or regulate release of soluble isoforms. In addition, it may alter the affinity of interaction with NKG2D or modulate cytotoxic activity from the target cell. In particular, ligands with high affinity for NKG2D are associated with down regulation of this protein on the effector cell, effectively limiting cytotoxic activity in a negative-feedback circuit. Given these observations, it is not surprising that NKG2DL alleles are associated with relative risk for development of several clinical disorders and the critical role of the NKG2D:NKG2DL interaction is demonstrated in many murine models. Increased understanding of the biophysical and functional consequences of this polymorphism is likely to provide insights into novel immunotherapeutic approaches.
Collapse
Affiliation(s)
- Jianmin Zuo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Fiyaz Mohammed
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Paul Moss
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
39
|
Böttcher K, Rombouts K, Saffioti F, Roccarina D, Rosselli M, Hall A, Luong T, Tsochatzis EA, Thorburn D, Pinzani M. MAIT cells are chronically activated in patients with autoimmune liver disease and promote profibrogenic hepatic stellate cell activation. Hepatology 2018; 68:172-186. [PMID: 29328499 DOI: 10.1002/hep.29782] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/13/2017] [Accepted: 01/09/2018] [Indexed: 12/12/2022]
Abstract
UNLABELLED Autoimmune liver diseases (AILDs) are chronic liver pathologies characterized by fibrosis and cirrhosis due to immune-mediated liver damage. In this study, we addressed the question whether mucosal-associated invariant T (MAIT) cells, innate-like T cells, are functionally altered in patients with AILD and whether MAIT cells can promote liver fibrosis through activation of hepatic stellate cells (HSCs). We analyzed the phenotype and function of MAIT cells from AILD patients and healthy controls by multicolor flow cytometry and investigated the interaction between human MAIT cells and primary human hepatic stellate cells (hHSCs). We show that MAIT cells are significantly decreased in peripheral blood and liver tissue of patients with AILD. Notably, MAIT cell frequency tended to decrease with increasing fibrosis stage. MAIT cells from AILD patients showed signs of exhaustion, such as impaired interferon-γ (IFN-γ) production and high ex vivo expression of the activation and exhaustion markers CD38, HLA-DR, and CTLA-4. Mechanistically, this exhausted state could be induced by repetitive stimulation of MAIT cells with the cytokines interleukin (IL)-12 and IL-18, leading to decreased IFN-γ and increased exhaustion marker expression. Of note, repetitive stimulation with IL-12 further resulted in expression of the profibrogenic cytokine IL-17A by otherwise exhausted MAIT cells. Accordingly, MAIT cells from both healthy controls and AILD patients were able to induce an activated, proinflammatory and profibrogenic phenotype in hHSCs in vitro that was partly mediated by IL-17. CONCLUSION Our data provide evidence that MAIT cells in AILD patients have evolved towards an exhausted, profibrogenic phenotype and can contribute to the development of HSC-mediated liver fibrosis. These findings reveal a cellular and molecular pathway for fibrosis development in AILD that could be exploited for antifibrotic therapy. (Hepatology 2018;68:172-186).
Collapse
Affiliation(s)
- Katrin Böttcher
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus London, United Kingdom.,Sheila Sherlock Liver Centre, Royal Free Hospital, London, United Kingdom
| | - Krista Rombouts
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus London, United Kingdom
| | - Francesca Saffioti
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus London, United Kingdom.,Sheila Sherlock Liver Centre, Royal Free Hospital, London, United Kingdom.,Department of Clinical and Experimental Medicine, Division of Clinical and Molecular Hepatology, University Hospital of Messina, Messina, Italy
| | - Davide Roccarina
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus London, United Kingdom.,Sheila Sherlock Liver Centre, Royal Free Hospital, London, United Kingdom
| | - Matteo Rosselli
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus London, United Kingdom.,Sheila Sherlock Liver Centre, Royal Free Hospital, London, United Kingdom
| | - Andrew Hall
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus London, United Kingdom
| | - TuVinh Luong
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus London, United Kingdom.,Sheila Sherlock Liver Centre, Royal Free Hospital, London, United Kingdom
| | - Emmanuel A Tsochatzis
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus London, United Kingdom.,Sheila Sherlock Liver Centre, Royal Free Hospital, London, United Kingdom
| | - Douglas Thorburn
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus London, United Kingdom.,Sheila Sherlock Liver Centre, Royal Free Hospital, London, United Kingdom
| | - Massimo Pinzani
- Regenerative Medicine and Fibrosis Group, Institute for Liver and Digestive Health, University College London, Royal Free Campus London, United Kingdom.,Sheila Sherlock Liver Centre, Royal Free Hospital, London, United Kingdom
| |
Collapse
|
40
|
Abstract
T cell receptors (TCRs) are protein complexes formed by six different polypeptides. In most T cells, TCRs are composed of αβ subunits displaying immunoglobulin-like variable domains that recognize peptide antigens associated with major histocompatibility complex molecules expressed on the surface of antigen-presenting cells. TCRαβ subunits are associated with the CD3 complex formed by the γ, δ, ε, and ζ subunits, which are invariable and ensure signal transduction. Here, we review how the expression and function of TCR complexes are orchestrated by several fine-tuned cellular processes that encompass (a) synthesis of the subunits and their correct assembly and expression at the plasma membrane as a single functional complex, (b) TCR membrane localization and dynamics at the plasma membrane and in endosomal compartments, (c) TCR signal transduction leading to T cell activation, and (d) TCR degradation. These processes balance each other to ensure efficient T cell responses to a variety of antigenic stimuli while preventing autoimmunity.
Collapse
Affiliation(s)
- Andrés Alcover
- Lymphocyte Cell Biology Unit, INSERM U1221, Department of Immunology, Institut Pasteur, Paris 75015, France; ,
| | - Balbino Alarcón
- Severo Ochoa Center for Molecular Biology, CSIC-UAM, Madrid 28049, Spain;
| | - Vincenzo Di Bartolo
- Lymphocyte Cell Biology Unit, INSERM U1221, Department of Immunology, Institut Pasteur, Paris 75015, France; ,
| |
Collapse
|
41
|
Iseka FM, Goetz BT, Mushtaq I, An W, Cypher LR, Bielecki TA, Tom EC, Arya P, Bhattacharyya S, Storck MD, Semerad CL, Talmadge JE, Mosley RL, Band V, Band H. Role of the EHD Family of Endocytic Recycling Regulators for TCR Recycling and T Cell Function. THE JOURNAL OF IMMUNOLOGY 2017; 200:483-499. [PMID: 29212907 DOI: 10.4049/jimmunol.1601793] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 11/01/2017] [Indexed: 12/31/2022]
Abstract
T cells use the endocytic pathway for key cell biological functions, including receptor turnover and maintenance of the immunological synapse. Some of the established players include the Rab GTPases, the SNARE complex proteins, and others, which function together with EPS-15 homology domain-containing (EHD) proteins in non-T cell systems. To date, the role of the EHD protein family in T cell function remains unexplored. We generated conditional EHD1/3/4 knockout mice using CD4-Cre and crossed these with mice bearing a myelin oligodendrocyte glycoprotein-specific TCR transgene. We found that CD4+ T cells from these mice exhibited reduced Ag-driven proliferation and IL-2 secretion in vitro. In vivo, these mice exhibited reduced severity of experimental autoimmune encephalomyelitis. Further analyses showed that recycling of the TCR-CD3 complex was impaired, leading to increased lysosomal targeting and reduced surface levels on CD4+ T cells of EHD1/3/4 knockout mice. Our studies reveal a novel role of the EHD family of endocytic recycling regulatory proteins in TCR-mediated T cell functions.
Collapse
Affiliation(s)
- Fany M Iseka
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198
| | - Benjamin T Goetz
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198
| | - Insha Mushtaq
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Wei An
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198
| | - Luke R Cypher
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198
| | - Timothy A Bielecki
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198
| | - Eric C Tom
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198
| | - Priyanka Arya
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198
| | - Sohinee Bhattacharyya
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Matthew D Storck
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198
| | - Craig L Semerad
- Flow Cytometry Research Facility, University of Nebraska Medical Center, Omaha, NE 68198; and
| | - James E Talmadge
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - R Lee Mosley
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198.,Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
| | - Vimla Band
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198
| | - Hamid Band
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198; .,Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198.,Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198.,Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
42
|
Hypercholesterolemia Enhances T Cell Receptor Signaling and Increases the Regulatory T Cell Population. Sci Rep 2017; 7:15655. [PMID: 29142309 PMCID: PMC5688061 DOI: 10.1038/s41598-017-15546-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 10/30/2017] [Indexed: 01/13/2023] Open
Abstract
Hypercholesterolemia promotes the inflammation against lipoproteins in atherosclerosis. Development of atherosclerosis is affected by the balance between pro-inflammatory effector T cells and anti-inflammatory regulatory T (Treg) cells. However, phenotype and function of T cell subpopulations in hypercholesterolemia remain to be investigated. Here, we found that cholesterol-containing diet increased the expression of the Treg cell lineage-defining transcription factor FoxP3 among thymocytes and splenocytes. Hypercholesterolemia elevated the FoxP3 expression level and population size of peripheral Treg cells, but did not prevent enhanced proliferation of stimulated T cells. Moreover, cholesterol supplementation in diet as well as in cell culture medium promoted T cell antigen receptor (TCR) signaling in CD4+ T cells. Our results demonstrate that hypercholesterolemia enhances TCR stimulation, Treg cell development as well as T cell proliferation. Thus, our findings may help to understand why hypercholesterolemia correlates with altered CD4+ T cell responses.
Collapse
|
43
|
Dar AA, Bhat SA, Gogoi D, Gokhale A, Chiplunkar SV. Inhibition of Notch signalling has ability to alter the proximal and distal TCR signalling events in human CD3 + αβ T-cells. Mol Immunol 2017; 92:116-124. [PMID: 29078088 DOI: 10.1016/j.molimm.2017.10.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 09/26/2017] [Accepted: 10/17/2017] [Indexed: 01/13/2023]
Abstract
The Notch signalling pathway is an important regulator of T cell function and is known to regulate the effector functions of T cells driven by T cell receptor (TCR). However, the mechanism integrating these pathways in human CD3+ αβ T cells is not well understood. The present study was carried out to investigate how Notch and TCR driven signalling are synchronized in human αβ T cells. Differential expression of Notch receptors, ligands, and target genes is observed on human αβ T cells which are upregulated on stimulation with α-CD3/CD28 mAb. Inhibition of Notch signalling by GSI-X inhibited the activation of T cells and affected proximal T cell signalling by regulating CD3-ζ chain expression. Inhibition of Notch signalling decreased the protein expression of CD3-ζ chain and induced expression of E3 ubiquitin ligase (GRAIL) in human αβ T cells. Apart from affecting proximal TCR signalling, Notch signalling also regulated the distal TCR signalling events. In the absence of Notch signalling, α-CD3/CD28 mAb induced activation and IFN-γ production by αβ T cells was down-modulated. The absence of Notch signalling in human αβ T cells inhibited proliferative responses despite strong signalling through TCR and IL-2 receptor. This study shows how Notch signalling cooperates with TCR signalling by regulating CD3-ζ chain expression to support proliferation and activation of human αβ T cells.
Collapse
Affiliation(s)
- Asif A Dar
- Chiplunkar Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra 410210, India; Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, Maharashtra 400094, India
| | - Sajad A Bhat
- Chiplunkar Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra 410210, India; Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, Maharashtra 400094, India
| | - Dimpu Gogoi
- Chiplunkar Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra 410210, India; Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, Maharashtra 400094, India
| | - Abhiram Gokhale
- Chiplunkar Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra 410210, India
| | - Shubhada V Chiplunkar
- Chiplunkar Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, Maharashtra 410210, India; Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai, Maharashtra 400094, India.
| |
Collapse
|
44
|
Zuo J, Willcox CR, Mohammed F, Davey M, Hunter S, Khan K, Antoun A, Katakia P, Croudace J, Inman C, Parry H, Briggs D, Malladi R, Willcox BE, Moss P. A disease-linked ULBP6 polymorphism inhibits NKG2D-mediated target cell killing by enhancing the stability of NKG2D ligand binding. Sci Signal 2017; 10:10/481/eaai8904. [PMID: 28559451 DOI: 10.1126/scisignal.aai8904] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
NKG2D (natural killer group 2, member D) is an activating receptor found on the surface of immune cells, including natural killer (NK) cells, which regulates innate and adaptive immunity through recognition of the stress-induced ligands ULBP1 (UL16 binding protein 1) to ULBP6 and MICA/B. Similar to class I human leukocyte antigen (HLA), these NKG2D ligands have a major histocompatibility complex-like fold and exhibit pronounced polymorphism, which influences human disease susceptibility. However, whereas class I HLA polymorphisms occur predominantly in the α1α2 groove and affect antigen binding, the effects of most NKG2D ligand polymorphisms are unclear. We studied the molecular and functional consequences of the two major alleles of ULBP6, the most polymorphic ULBP gene, which are associated with autoimmunity and relapse after stem cell transplantation. Surface plasmon resonance and crystallography studies revealed that the arginine-to-leucine polymorphism within ULBP0602 affected the NKG2D-ULBP6 interaction by generating an energetic hotspot. This resulted in an NKG2D-ULBP0602 affinity of 15.5 nM, which is 10- to 1000-fold greater than the affinities of other ULBP-NKG2D interactions and limited NKG2D-mediated activation. In addition, soluble ULBP0602 exhibited high-affinity competitive binding for NKG2D and partially suppressed NKG2D-mediated activation of NK cells by other NKG2D ligands. These effects resulted in a decrease in a range of NKG2D-mediated effector functions. Our results reveal that ULBP polymorphisms affect the strength of human lymphocyte responses to cellular stress signals and may offer opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Jianmin Zuo
- Cancer Immunology and Immunotherapy Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Vincent Drive, Edgbaston, Birmingham B15 2TT, UK
| | - Carrie R Willcox
- Cancer Immunology and Immunotherapy Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Vincent Drive, Edgbaston, Birmingham B15 2TT, UK
| | - Fiyaz Mohammed
- Cancer Immunology and Immunotherapy Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Vincent Drive, Edgbaston, Birmingham B15 2TT, UK
| | - Martin Davey
- Cancer Immunology and Immunotherapy Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Vincent Drive, Edgbaston, Birmingham B15 2TT, UK
| | - Stuart Hunter
- Cancer Immunology and Immunotherapy Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Vincent Drive, Edgbaston, Birmingham B15 2TT, UK
| | - Kabir Khan
- Cancer Immunology and Immunotherapy Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Vincent Drive, Edgbaston, Birmingham B15 2TT, UK
| | - Ayman Antoun
- Cancer Immunology and Immunotherapy Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Vincent Drive, Edgbaston, Birmingham B15 2TT, UK
| | - Poonam Katakia
- Cancer Immunology and Immunotherapy Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Vincent Drive, Edgbaston, Birmingham B15 2TT, UK
| | - Joanne Croudace
- Cancer Immunology and Immunotherapy Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Vincent Drive, Edgbaston, Birmingham B15 2TT, UK
| | - Charlotte Inman
- Cancer Immunology and Immunotherapy Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Vincent Drive, Edgbaston, Birmingham B15 2TT, UK
| | - Helen Parry
- Cancer Immunology and Immunotherapy Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Vincent Drive, Edgbaston, Birmingham B15 2TT, UK.,Department of Haematology, University Hospital Birmingham, Birmingham B15 2TH, UK
| | - David Briggs
- National Health Service Blood and Transplant, Birmingham B15 2SG, UK
| | - Ram Malladi
- Cancer Immunology and Immunotherapy Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Vincent Drive, Edgbaston, Birmingham B15 2TT, UK.,Department of Haematology, University Hospital Birmingham, Birmingham B15 2TH, UK
| | - Benjamin E Willcox
- Cancer Immunology and Immunotherapy Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Vincent Drive, Edgbaston, Birmingham B15 2TT, UK.
| | - Paul Moss
- Cancer Immunology and Immunotherapy Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Vincent Drive, Edgbaston, Birmingham B15 2TT, UK. .,Department of Haematology, University Hospital Birmingham, Birmingham B15 2TH, UK
| |
Collapse
|
45
|
Balyan R, Gund R, Ebenezer C, Khalsa JK, Verghese DA, Krishnamurthy T, George A, Bal V, Rath S, Chaudhry A. Modulation of Naive CD8 T Cell Response Features by Ligand Density, Affinity, and Continued Signaling via Internalized TCRs. THE JOURNAL OF IMMUNOLOGY 2017; 198:1823-1837. [PMID: 28100678 DOI: 10.4049/jimmunol.1600083] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 12/20/2016] [Indexed: 01/08/2023]
Abstract
T cell response magnitudes increase with increasing antigenic dosage. However, it is unclear whether ligand density only modulates the proportions of responding ligand-specific T cells or also alters responses at the single cell level. Using brief (3 h) exposure of TCR-transgenic mouse CD8 T cells in vitro to varying densities of cognate peptide-MHC ligand followed by ligand-free culture in IL-2, we found that ligand density determined the frequencies of responding cells but not the expression levels of the early activation marker molecule, CD69. Cells with low glucose uptake capacity and low protein synthesis rates were less ligand-sensitive, implicating metabolic competence in the response heterogeneity of CD8 T cell populations. Although most responding cells proliferated, ligand density was associated with time of entry into proliferation and with the extent of cell surface TCR downmodulation. TCR internalization was associated, regardless of the ligand density, with rapidity of c-myc induction, loss of the cell cycle inhibitor p27kip1, metabolic reprogramming, and cell cycle entry. A low affinity peptide ligand behaved, regardless of ligand density, like a low density, high affinity ligand in all these parameters. Inhibition of signaling after ligand exposure selectively delayed proliferation in cells with internalized TCRs. Finally, internalized TCRs continued to signal and genetic modification of TCR internalization and trafficking altered the duration of signaling in a T cell hybridoma. Together, our findings indicate that heterogeneity among responding CD8 T cell populations in their ability to respond to TCR-mediated stimulation and internalize TCRs mediates detection of ligand density or affinity, contributing to graded response magnitudes.
Collapse
Affiliation(s)
- Renu Balyan
- National Institute of Immunology, New Delhi 110067, India; and
| | - Rupali Gund
- National Institute of Immunology, New Delhi 110067, India; and
| | - Chitra Ebenezer
- National Institute of Immunology, New Delhi 110067, India; and
| | | | | | | | - Anna George
- National Institute of Immunology, New Delhi 110067, India; and
| | - Vineeta Bal
- National Institute of Immunology, New Delhi 110067, India; and
| | - Satyajit Rath
- National Institute of Immunology, New Delhi 110067, India; and
| | - Ashutosh Chaudhry
- National Institute of Immunology, New Delhi 110067, India; and.,Memorial Sloan Kettering Cancer Center, New York, NY 10065
| |
Collapse
|
46
|
Abstract
Mast cells are key effector cells in inflammation that can be activated by specific antigens via IgE or IgG binding on their FcR. Aggregation of mast cell Fc receptors by cell-bound antigens induces mast cell polarized degranulation toward the stimulatory cell, a process named antibody-dependent degranulatory synapse (ADDS). This polarized degranulation allows mast cells to expose bioactive material embedded in the granule matrix toward the antibody-targeted cell and is accompanied by the formation of a signaling area at the cell-cell contact site. In this chapter, we describe (1) how to stimulate mast cells with cell-bound antigens and (2) how to monitor ADDS formation and to investigate ADDS characteristics by confocal microscopy.
Collapse
|
47
|
Apinun J, Sengprasert P, Yuktanandana P, Ngarmukos S, Tanavalee A, Reantragoon R. Immune Mediators in Osteoarthritis: Infrapatellar Fat Pad-Infiltrating CD8+ T Cells Are Increased in Osteoarthritic Patients with Higher Clinical Radiographic Grading. Int J Rheumatol 2016; 2016:9525724. [PMID: 28070192 PMCID: PMC5192329 DOI: 10.1155/2016/9525724] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 10/25/2016] [Accepted: 11/09/2016] [Indexed: 01/15/2023] Open
Abstract
Osteoarthritis is a condition of joint failure characterized by many pathologic changes of joint-surrounding tissues. Many evidences suggest the role of both innate and adaptive immunity that interplay, resulting either in initiation or in progression of osteoarthritis. Adaptive immune cells, in particular T cells, have been demonstrated to play a role in the development of OA in animal models. However, the underlying mechanism is yet unclear. Our aim was to correlate the frequency and phenotype of tissue-infiltrating T cells in the synovial tissue and infrapatellar fat pad with radiographic grading. Our results show that CD8+ T cells are increased in osteoarthritic patients with higher radiographic grading. When peripheral blood CD8+ T cells were examined, we show that CD8+ T cells possess a significantly higher level of activation than its CD4+ T cell counterpart (P < 0.0001). Our results suggest a role for CD8+ T cells and recruitment of these activated circulating peripheral blood CD8+ T cells to the knee triggering local inflammation within the knee joint.
Collapse
Affiliation(s)
- Jirun Apinun
- Department of Orthopedics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Panjana Sengprasert
- Immunology Division, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Pongsak Yuktanandana
- Department of Orthopedics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Srihatach Ngarmukos
- Department of Orthopedics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Aree Tanavalee
- Department of Orthopedics, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Rangsima Reantragoon
- Immunology Division, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Immunology and Immune-Mediated Diseases, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
48
|
Ali Q, Mustafa IH, Elkamel A, Touboul E, Gruy F, Lambert C. Mathematical modelling of T-cells activation dynamics for CD3 molecules recycling process. CAN J CHEM ENG 2016. [DOI: 10.1002/cjce.22607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Qasim Ali
- Department of Applied Mathematics; University of Western Ontario; London ON Canada
| | - Ibrahim H. Mustafa
- Biomedical Engineering Department; Helwan University; Cairo Egypt
- Department of Chemical Engineering; University of Waterloo; Waterloo ON Canada
| | - Ali Elkamel
- Department of Chemical Engineering; University of Waterloo; Waterloo ON Canada
| | - Eric Touboul
- Henry Fayol Institute; ENSM - Saint Étienne France
| | - Frederic Gruy
- Center for Biomedical and Healthcare Engineering; ENSM - Saint Étienne France
| | - Claude Lambert
- CHU - Saint Etienne, Immunology Lab; University Hospital; F 42055, Saint-Étienne France
| |
Collapse
|
49
|
Jones RB, Mueller S, Kumari S, Vrbanac V, Genel S, Tager AM, Allen TM, Walker BD, Irvine DJ. Antigen recognition-triggered drug delivery mediated by nanocapsule-functionalized cytotoxic T-cells. Biomaterials 2016; 117:44-53. [PMID: 27936416 DOI: 10.1016/j.biomaterials.2016.11.048] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 11/20/2016] [Accepted: 11/24/2016] [Indexed: 01/23/2023]
Abstract
Cytotoxic T-Lymphocytes (CTLs) kill pathogen-infected or transformed cells following interaction of their T-cell receptors (TCRs) with foreign (e.g. virus-derived) peptides bound to MHC-I molecules on the target cell. TCR binding triggers CTLs to secrete perforin, which forms pores in the target cell membrane, promoting target death. Here, we show that by conjugating drug-loaded lipid nanoparticles to the surface of CTLs, their lytic machinery can be co-opted to lyse the cell-bound drug carrier, providing triggered release of drug cargo upon target cell recognition. Protein encapsulated in T-cell-bound nanoparticles was released following culture of CTLs with target cells in an antigen dose- and perforin-dependent manner and coincided with target cell lysis. Using this approach, we demonstrate the capacity of HIV-specific CTLs to deliver an immunotherapeutic agent to an anatomical site of viral replication. This strategy provides a novel means to couple drug delivery to the action of therapeutic cells in vivo.
Collapse
Affiliation(s)
- R Brad Jones
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Dept of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington DC, USA; Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
| | - Stephanie Mueller
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
| | - Sudha Kumari
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
| | - Vlad Vrbanac
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Center for Immunology and Inflammatory Diseases, MGH, Boston, MA, USA
| | - Shy Genel
- Astronomy Dept., Columbia University, New York, NY, USA
| | - Andrew M Tager
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Todd M Allen
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Bruce D Walker
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Dept. of Microbiology and Immunology, Harvard Medical School, Boston, MA, USA; Dept. of Biological Engineering, MIT, Cambridge, MA, USA; Dept. of Materials Science & Engineering, MIT, Cambridge, MA, USA
| | - Darrell J Irvine
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA; Dept. of Biological Engineering, MIT, Cambridge, MA, USA; Dept. of Materials Science & Engineering, MIT, Cambridge, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
50
|
Surface-bound bovine serum albumin carrier protein as present in recombinant cytokine preparations amplifies T helper 17 cell polarization. Sci Rep 2016; 6:36598. [PMID: 27808281 PMCID: PMC5093436 DOI: 10.1038/srep36598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 10/17/2016] [Indexed: 11/18/2022] Open
Abstract
Understanding of T helper 17 lineage (TH17) polarization has been significantly promoted by cell culture experiments that reduce the complexity of the in vivo environment. We here investigated TH17 amplification by coating of cytokine preparations. Cytokine preparations coated to the surface compared to the same amount given in solution significantly enhanced TH17 polarization assessed by flow cytometry and interleukin (IL)-17A, IL-17F and RORγt mRNA expression. T cell proliferation and TH1 polarization were similarly enhanced while TREG polarization was impeded. TH17 amplification was replicated by coating the plate with low amounts of FCS or albumin as used as carrier protein for cytokines (0.5 μl 0.1%). It was unaltered by filtration, protein digestion and arylhydrocarbon receptor blockade, not replicated by LPS and independent of integrin stimulation. TH17 amplification required anti-CD3 stimulation and was T cell intrinsic. Supernatants of CD4+ cells polarized on coated cytokine preparations with carrier albumin conferred amplification to fresh splenocytes. Coating markedly elevated CD4+ IL-22 mRNA expression and IL-22 blockade significantly reduced TH17 amplification. Our data show TH17 amplification by coated albumin in the low amounts present in recombinant cytokine preparations. This unexpected adjuvant like effect underscores the need for controls also for temporal and spatial factors in cell culture.
Collapse
|