Skip to main content
Top
Published in: Cancer Immunology, Immunotherapy 6/2023

Open Access 23-12-2022 | Graft-Versus-Host Disease | Research

TCF-1 regulates NKG2D expression on CD8 T cells during anti-tumor responses

Authors: Rebecca Harris, Mahinbanu Mammadli, Shannon Hiner, Liye Suo, Qi Yang, Jyoti Misra Sen, Mobin Karimi

Published in: Cancer Immunology, Immunotherapy | Issue 6/2023

Login to get access

Abstract

Cancer immunotherapy relies on improving T cell effector functions against malignancies, but despite the identification of several key transcription factors (TFs), the biological functions of these TFs are not entirely understood. We developed and utilized a novel, clinically relevant murine model to dissect the functional properties of crucial T cell transcription factors during anti-tumor responses. Our data showed that the loss of TCF-1 in CD8 T cells also leads to loss of key stimulatory molecules such as CD28. Our data showed that TCF-1 suppresses surface NKG2D expression on naïve and activated CD8 T cells via key transcriptional factors Eomes and T-bet. Using both in vitro and in vivo models, we uncovered how TCF-1 regulates critical molecules responsible for peripheral CD8 T cell effector functions. Finally, our unique genetic and molecular approaches suggested that TCF-1 also differentially regulates essential kinases. These kinases, including LCK, LAT, ITK, PLC-γ1, P65, ERKI/II, and JAK/STATs, are required for peripheral CD8 T cell persistent function during alloimmunity. Overall, our molecular and bioinformatics data demonstrate the mechanism by which TCF-1 modulated several critical aspects of T cell function during CD8 T cell response to cancer.

Graphical abstract

Summary Figure: TCF-1 is required for persistent function of CD8 T cells but dispensable for anti-tumor response. Here, we have utilized a novel mouse model that lacks TCF-1 specifically on CD8 T cells for an allogeneic transplant model. We uncovered a molecular mechanism of how TCF-1 regulates key signaling pathways at both transcriptomic and protein levels. These key molecules included LCK, LAT, ITK, PLC-γ1, p65, ERK I/II, and JAK/STAT signaling. Next, we showed that the lack of TCF-1 impacted phenotype, proinflammatory cytokine production, chemokine expression, and T cell activation. We provided clinical evidence for how these changes impact GVHD target organs (skin, small intestine, and liver). Finally, we provided evidence that TCF-1 regulates NKG2D expression on mouse naïve and activated CD8 T cells. We have shown that CD8 T cells from TCF-1 cKO mice mediate cytolytic functions via NKG2D.
Appendix
Available only for authorised users
Literature
5.
go back to reference Germar K et al (2011) T-cell factor 1 is a gatekeeper for T-cell specification in response to Notch signaling. Proc Natl Acad Sci U S A 108(50):20060–20065PubMedPubMedCentralCrossRef Germar K et al (2011) T-cell factor 1 is a gatekeeper for T-cell specification in response to Notch signaling. Proc Natl Acad Sci U S A 108(50):20060–20065PubMedPubMedCentralCrossRef
7.
go back to reference Beltra JC et al (2020) Developmental relationships of four exhausted CD8(+) T cell subsets reveals underlying transcriptional and epigenetic landscape control mechanisms. Immunity 52(5):825-841 e8PubMedPubMedCentralCrossRef Beltra JC et al (2020) Developmental relationships of four exhausted CD8(+) T cell subsets reveals underlying transcriptional and epigenetic landscape control mechanisms. Immunity 52(5):825-841 e8PubMedPubMedCentralCrossRef
8.
go back to reference Zander R et al (2019) CD4(+) T cell help is required for the formation of a cytolytic CD8(+) T cell subset that protects against chronic infection and cancer. Immunity 51(6):1028-1042 e4PubMedPubMedCentralCrossRef Zander R et al (2019) CD4(+) T cell help is required for the formation of a cytolytic CD8(+) T cell subset that protects against chronic infection and cancer. Immunity 51(6):1028-1042 e4PubMedPubMedCentralCrossRef
9.
go back to reference LaFleur MW et al (2019) PTPN2 regulates the generation of exhausted CD8(+) T cell subpopulations and restrains tumor immunity. Nat Immunol 20(10):1335–1347PubMedPubMedCentralCrossRef LaFleur MW et al (2019) PTPN2 regulates the generation of exhausted CD8(+) T cell subpopulations and restrains tumor immunity. Nat Immunol 20(10):1335–1347PubMedPubMedCentralCrossRef
10.
go back to reference Kurtulus S et al (2019) Checkpoint blockade immunotherapy induces dynamic changes in PD-1(-)CD8(+) tumor-infiltrating T cells. Immunity 50(1):181-194 e6PubMedPubMedCentralCrossRef Kurtulus S et al (2019) Checkpoint blockade immunotherapy induces dynamic changes in PD-1(-)CD8(+) tumor-infiltrating T cells. Immunity 50(1):181-194 e6PubMedPubMedCentralCrossRef
11.
go back to reference Siddiqui I et al (2019) Intratumoral Tcf1(+)PD-1(+)CD8(+) T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy. Immunity 50(1):195-211 e10PubMedCrossRef Siddiqui I et al (2019) Intratumoral Tcf1(+)PD-1(+)CD8(+) T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy. Immunity 50(1):195-211 e10PubMedCrossRef
12.
go back to reference Utzschneider DT et al (2013) T cells maintain an exhausted phenotype after antigen withdrawal and population reexpansion. Nat Immunol 14(6):603–610PubMedCrossRef Utzschneider DT et al (2013) T cells maintain an exhausted phenotype after antigen withdrawal and population reexpansion. Nat Immunol 14(6):603–610PubMedCrossRef
13.
go back to reference Schietinger A et al (2016) Tumor-specific T cell dysfunction is a dynamic antigen-driven differentiation program initiated early during tumorigenesis. Immunity 45(2):389–401PubMedPubMedCentralCrossRef Schietinger A et al (2016) Tumor-specific T cell dysfunction is a dynamic antigen-driven differentiation program initiated early during tumorigenesis. Immunity 45(2):389–401PubMedPubMedCentralCrossRef
15.
18.
go back to reference Lugli E et al (2013) Superior T memory stem cell persistence supports long-lived T cell memory. J Clin Invest 123(2):594–599PubMedPubMedCentral Lugli E et al (2013) Superior T memory stem cell persistence supports long-lived T cell memory. J Clin Invest 123(2):594–599PubMedPubMedCentral
19.
go back to reference Cieri N et al (2013) IL-7 and IL-15 instruct the generation of human memory stem T cells from naive precursors. Blood 121(4):573–584PubMedCrossRef Cieri N et al (2013) IL-7 and IL-15 instruct the generation of human memory stem T cells from naive precursors. Blood 121(4):573–584PubMedCrossRef
20.
go back to reference Cui W et al (2011) An interleukin-21-interleukin-10-STAT3 pathway is critical for functional maturation of memory CD8+ T cells. Immunity 35(5):792–805PubMedPubMedCentralCrossRef Cui W et al (2011) An interleukin-21-interleukin-10-STAT3 pathway is critical for functional maturation of memory CD8+ T cells. Immunity 35(5):792–805PubMedPubMedCentralCrossRef
21.
go back to reference Balassa K, Danby R, Rocha V (2019) Haematopoietic stem cell transplants: principles and indications. Br J Hosp Med (Lond) 80(1):33–39PubMedCrossRef Balassa K, Danby R, Rocha V (2019) Haematopoietic stem cell transplants: principles and indications. Br J Hosp Med (Lond) 80(1):33–39PubMedCrossRef
23.
go back to reference Hall E, Shenoy S (2019) Hematopoietic stem cell transplantation: a neonatal perspective. NeoReviews 20(6):e336–e345PubMedCrossRef Hall E, Shenoy S (2019) Hematopoietic stem cell transplantation: a neonatal perspective. NeoReviews 20(6):e336–e345PubMedCrossRef
25.
26.
go back to reference Wang Y et al (2019) The transcription factor TCF1 preserves the effector function of exhausted CD8 T cells during chronic viral infection. Front Immunol 10:169PubMedPubMedCentralCrossRef Wang Y et al (2019) The transcription factor TCF1 preserves the effector function of exhausted CD8 T cells during chronic viral infection. Front Immunol 10:169PubMedPubMedCentralCrossRef
27.
29.
30.
33.
go back to reference Dutt S et al (2011) CD8+CD44(hi) but not CD4+CD44(hi) memory T cells mediate potent graft antilymphoma activity without GVHD. Blood 117(11):3230–3239PubMedPubMedCentralCrossRef Dutt S et al (2011) CD8+CD44(hi) but not CD4+CD44(hi) memory T cells mediate potent graft antilymphoma activity without GVHD. Blood 117(11):3230–3239PubMedPubMedCentralCrossRef
34.
go back to reference Huang W et al (2019) Donor Allospecific CD44(high) central memory T cells have decreased ability to mediate graft-vs.-host disease. Front Immunol 10:624PubMedPubMedCentralCrossRef Huang W et al (2019) Donor Allospecific CD44(high) central memory T cells have decreased ability to mediate graft-vs.-host disease. Front Immunol 10:624PubMedPubMedCentralCrossRef
35.
go back to reference Mammadli M et al (2021) Interleukin-2-inducible T-cell kinase (Itk) signaling regulates potent noncanonical regulatory T cells. Clin Transl Med 11(12):e625PubMedPubMedCentralCrossRef Mammadli M et al (2021) Interleukin-2-inducible T-cell kinase (Itk) signaling regulates potent noncanonical regulatory T cells. Clin Transl Med 11(12):e625PubMedPubMedCentralCrossRef
40.
go back to reference Stojanovic A, Correia MP, Cerwenka A (2018) The NKG2D/NKG2DL axis in the crosstalk between lymphoid and myeloid cells in health and disease. Front Immunol 9:827PubMedPubMedCentralCrossRef Stojanovic A, Correia MP, Cerwenka A (2018) The NKG2D/NKG2DL axis in the crosstalk between lymphoid and myeloid cells in health and disease. Front Immunol 9:827PubMedPubMedCentralCrossRef
41.
go back to reference Hu J et al (2016) Regulation of NKG2D(+)CD8(+) T-cell-mediated antitumor immune surveillance: identification of a novel CD28 activation-mediated, STAT3 phosphorylation-dependent mechanism. Oncoimmunology 5(12):e1252012PubMedPubMedCentralCrossRef Hu J et al (2016) Regulation of NKG2D(+)CD8(+) T-cell-mediated antitumor immune surveillance: identification of a novel CD28 activation-mediated, STAT3 phosphorylation-dependent mechanism. Oncoimmunology 5(12):e1252012PubMedPubMedCentralCrossRef
42.
go back to reference Maasho K et al (2005) NKG2D is a costimulatory receptor for human naive CD8+ T cells. J Immunol 174(8):4480–4484PubMedCrossRef Maasho K et al (2005) NKG2D is a costimulatory receptor for human naive CD8+ T cells. J Immunol 174(8):4480–4484PubMedCrossRef
43.
46.
go back to reference Steinke FC et al (2014) TCF-1 and LEF-1 act upstream of Th-POK to promote the CD4(+) T cell fate and interact with Runx3 to silence Cd4 in CD8(+) T cells. Nat Immunol 15(7):646–656PubMedPubMedCentralCrossRef Steinke FC et al (2014) TCF-1 and LEF-1 act upstream of Th-POK to promote the CD4(+) T cell fate and interact with Runx3 to silence Cd4 in CD8(+) T cells. Nat Immunol 15(7):646–656PubMedPubMedCentralCrossRef
47.
go back to reference Edinger M et al (2003) CD4+CD25+ regulatory T cells preserve graft-versus-tumor activity while inhibiting graft-versus-host disease after bone marrow transplantation. Nat Med 9(9):1144–1150PubMedCrossRef Edinger M et al (2003) CD4+CD25+ regulatory T cells preserve graft-versus-tumor activity while inhibiting graft-versus-host disease after bone marrow transplantation. Nat Med 9(9):1144–1150PubMedCrossRef
48.
49.
go back to reference Karimi MA et al (2014) A truncated human NKG2D splice isoform negatively regulates NKG2D-mediated function. J Immunol 193(6):2764–2771PubMedCrossRef Karimi MA et al (2014) A truncated human NKG2D splice isoform negatively regulates NKG2D-mediated function. J Immunol 193(6):2764–2771PubMedCrossRef
50.
go back to reference Karimi MA et al (2015) NKG2D expression by CD8+ T cells contributes to GVHD and GVT effects in a murine model of allogeneic HSCT. Blood 125(23):3655–3663PubMedPubMedCentralCrossRef Karimi MA et al (2015) NKG2D expression by CD8+ T cells contributes to GVHD and GVT effects in a murine model of allogeneic HSCT. Blood 125(23):3655–3663PubMedPubMedCentralCrossRef
51.
go back to reference Bray NL et al (2016) Near-optimal probabilistic RNA-seq quantification. Nat Biotechnol 34(5):525–527PubMedCrossRef Bray NL et al (2016) Near-optimal probabilistic RNA-seq quantification. Nat Biotechnol 34(5):525–527PubMedCrossRef
53.
54.
55.
go back to reference Berga-Bolanos R et al (2015) Cell-autonomous requirement for TCF1 and LEF1 in the development of Natural Killer T cells. Mol Immunol 68(2 Pt B):484–489PubMedPubMedCentralCrossRef Berga-Bolanos R et al (2015) Cell-autonomous requirement for TCF1 and LEF1 in the development of Natural Killer T cells. Mol Immunol 68(2 Pt B):484–489PubMedPubMedCentralCrossRef
56.
go back to reference Edinger M et al (2003) Evaluation of effector cell fate and function by in vivo bioluminescence imaging. Methods 31(2):172–179PubMedCrossRef Edinger M et al (2003) Evaluation of effector cell fate and function by in vivo bioluminescence imaging. Methods 31(2):172–179PubMedCrossRef
57.
go back to reference Hoffmann P et al (2002) Donor-type CD4(+)CD25(+) regulatory T cells suppress lethal acute graft-versus-host disease after allogeneic bone marrow transplantation. J Exp Med 196(3):389–399PubMedPubMedCentralCrossRef Hoffmann P et al (2002) Donor-type CD4(+)CD25(+) regulatory T cells suppress lethal acute graft-versus-host disease after allogeneic bone marrow transplantation. J Exp Med 196(3):389–399PubMedPubMedCentralCrossRef
60.
go back to reference Decman V et al (2010) Cell-intrinsic defects in the proliferative response of antiviral memory CD8 T cells in aged mice upon secondary infection. J Immunol 184(9):5151–5159PubMedCrossRef Decman V et al (2010) Cell-intrinsic defects in the proliferative response of antiviral memory CD8 T cells in aged mice upon secondary infection. J Immunol 184(9):5151–5159PubMedCrossRef
61.
go back to reference Zhu Y et al (2010) T-bet and eomesodermin are required for T cell-mediated antitumor immune responses. J Immunol 185(6):3174–3183PubMedCrossRef Zhu Y et al (2010) T-bet and eomesodermin are required for T cell-mediated antitumor immune responses. J Immunol 185(6):3174–3183PubMedCrossRef
62.
go back to reference Pais Ferreira D et al (2020) Central memory CD8(+) T cells derive from stem-like Tcf7(hi) effector cells in the absence of cytotoxic differentiation. Immunity 53(5):985-1000 e11PubMedCrossRef Pais Ferreira D et al (2020) Central memory CD8(+) T cells derive from stem-like Tcf7(hi) effector cells in the absence of cytotoxic differentiation. Immunity 53(5):985-1000 e11PubMedCrossRef
63.
go back to reference Williams JA et al (2005) Regulated costimulation in the thymus is critical for T cell development: dysregulated CD28 costimulation can bypass the pre-TCR checkpoint. J Immunol 175(7):4199–4207PubMedCrossRef Williams JA et al (2005) Regulated costimulation in the thymus is critical for T cell development: dysregulated CD28 costimulation can bypass the pre-TCR checkpoint. J Immunol 175(7):4199–4207PubMedCrossRef
65.
go back to reference Linsley PS, Ledbetter JA (1993) The role of the CD28 receptor during T cell responses to antigen. Annu Rev Immunol 11:191–212PubMedCrossRef Linsley PS, Ledbetter JA (1993) The role of the CD28 receptor during T cell responses to antigen. Annu Rev Immunol 11:191–212PubMedCrossRef
66.
go back to reference Borowski AB et al (2007) Memory CD8+ T cells require CD28 costimulation. J Immunol 179(10):6494–6503PubMedCrossRef Borowski AB et al (2007) Memory CD8+ T cells require CD28 costimulation. J Immunol 179(10):6494–6503PubMedCrossRef
67.
go back to reference Maly K, Schirmer M (2015) The story of CD4+ CD28- T cells revisited: Solved or still ongoing? J Immunol Res 2015:348746PubMedPubMedCentral Maly K, Schirmer M (2015) The story of CD4+ CD28- T cells revisited: Solved or still ongoing? J Immunol Res 2015:348746PubMedPubMedCentral
68.
72.
go back to reference Raulet DH (2003) Roles of the NKG2D immunoreceptor and its ligands. Nat Rev Immunol 3(10):781–790PubMedCrossRef Raulet DH (2003) Roles of the NKG2D immunoreceptor and its ligands. Nat Rev Immunol 3(10):781–790PubMedCrossRef
73.
go back to reference Chu T et al (2013) Bystander-activated memory CD8 T cells control early pathogen load in an innate-like, NKG2D-dependent manner. Cell Rep 3(3):701–708PubMedPubMedCentralCrossRef Chu T et al (2013) Bystander-activated memory CD8 T cells control early pathogen load in an innate-like, NKG2D-dependent manner. Cell Rep 3(3):701–708PubMedPubMedCentralCrossRef
74.
go back to reference Liu L et al (2022) The expression and function of NK cells in patients with acute myeloid leukemia. Zhongguo Shi Yan Xue Ye Xue Za Zhi 30(1):49–55PubMed Liu L et al (2022) The expression and function of NK cells in patients with acute myeloid leukemia. Zhongguo Shi Yan Xue Ye Xue Za Zhi 30(1):49–55PubMed
75.
go back to reference Wang S et al (2022) Blocking CD47 promotes anti-tumor immunity through CD103+ dendritic cell-NK cell axis in murine hepatocellular carcinoma model. J Hepatol 77:467PubMedCrossRef Wang S et al (2022) Blocking CD47 promotes anti-tumor immunity through CD103+ dendritic cell-NK cell axis in murine hepatocellular carcinoma model. J Hepatol 77:467PubMedCrossRef
76.
go back to reference Nishimura R et al (2008) In vivo trafficking and survival of cytokine-induced killer cells resulting in minimal GVHD with retention of antitumor activity. Blood 112(6):2563–2574PubMedPubMedCentralCrossRef Nishimura R et al (2008) In vivo trafficking and survival of cytokine-induced killer cells resulting in minimal GVHD with retention of antitumor activity. Blood 112(6):2563–2574PubMedPubMedCentralCrossRef
77.
go back to reference Jones AB et al (2022) Regulation of NKG2D stress ligands and its relevance in cancer progression. Cancers (Basel) 14(9):2339PubMedCrossRef Jones AB et al (2022) Regulation of NKG2D stress ligands and its relevance in cancer progression. Cancers (Basel) 14(9):2339PubMedCrossRef
78.
go back to reference Chan JK et al (2006) Enhanced killing of primary ovarian cancer by retargeting autologous cytokine-induced killer cells with bispecific antibodies: a preclinical study. Clin Cancer Res 12(6):1859–1867PubMedCrossRef Chan JK et al (2006) Enhanced killing of primary ovarian cancer by retargeting autologous cytokine-induced killer cells with bispecific antibodies: a preclinical study. Clin Cancer Res 12(6):1859–1867PubMedCrossRef
79.
go back to reference Karimi M et al (2005) Silencing human NKG2D, DAP10, and DAP12 reduces cytotoxicity of activated CD8+ T cells and NK cells. J Immunol 175(12):7819–7828PubMedCrossRef Karimi M et al (2005) Silencing human NKG2D, DAP10, and DAP12 reduces cytotoxicity of activated CD8+ T cells and NK cells. J Immunol 175(12):7819–7828PubMedCrossRef
80.
go back to reference Verneris MR et al (2004) Role of NKG2D signaling in the cytotoxicity of activated and expanded CD8+ T cells. Blood 103(8):3065–3072PubMedCrossRef Verneris MR et al (2004) Role of NKG2D signaling in the cytotoxicity of activated and expanded CD8+ T cells. Blood 103(8):3065–3072PubMedCrossRef
81.
go back to reference Lynch Kelly D et al (2015) Symptoms, cytokines, and quality of life in patients diagnosed with chronic graft-versus-host disease following allogeneic hematopoietic stem cell transplantation. Oncol Nurs Forum 42(3):265–275PubMedCrossRef Lynch Kelly D et al (2015) Symptoms, cytokines, and quality of life in patients diagnosed with chronic graft-versus-host disease following allogeneic hematopoietic stem cell transplantation. Oncol Nurs Forum 42(3):265–275PubMedCrossRef
82.
go back to reference D’Aveni M et al (2015) G-CSF mobilizes CD34+ regulatory monocytes that inhibit graft-versus-host disease. Sci Transl Med 7(281):281ra42PubMed D’Aveni M et al (2015) G-CSF mobilizes CD34+ regulatory monocytes that inhibit graft-versus-host disease. Sci Transl Med 7(281):281ra42PubMed
83.
go back to reference Beilhack A et al (2005) In vivo analyses of early events in acute graft-versus-host disease reveal sequential infiltration of T-cell subsets. Blood 106(3):1113–1122PubMedPubMedCentralCrossRef Beilhack A et al (2005) In vivo analyses of early events in acute graft-versus-host disease reveal sequential infiltration of T-cell subsets. Blood 106(3):1113–1122PubMedPubMedCentralCrossRef
84.
go back to reference Ferrara JL (2014) Blood and marrow transplant clinical trials network: progress since the state of the science symposium 2007. Biol Blood Marrow Transpl 20(2):149–153CrossRef Ferrara JL (2014) Blood and marrow transplant clinical trials network: progress since the state of the science symposium 2007. Biol Blood Marrow Transpl 20(2):149–153CrossRef
86.
go back to reference Blessin NC et al (2021) Prognostic role of proliferating CD8(+) cytotoxic Tcells in human cancers. Cell Oncol (Dordr) 44(4):793–803PubMedCrossRef Blessin NC et al (2021) Prognostic role of proliferating CD8(+) cytotoxic Tcells in human cancers. Cell Oncol (Dordr) 44(4):793–803PubMedCrossRef
90.
go back to reference da Huang W, Sherman BT, Lempicki RA (2009) Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 4(1):44–57PubMedCrossRef da Huang W, Sherman BT, Lempicki RA (2009) Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 4(1):44–57PubMedCrossRef
92.
go back to reference He R et al (2016) Follicular CXCR5- expressing CD8(+) T cells curtail chronic viral infection. Nature 537(7620):412–428PubMedCrossRef He R et al (2016) Follicular CXCR5- expressing CD8(+) T cells curtail chronic viral infection. Nature 537(7620):412–428PubMedCrossRef
93.
go back to reference Utzschneider DT et al (2016) T cell factor 1-expressing memory-like CD8(+) T cells sustain the immune response to chronic viral infections. Immunity 45(2):415–427PubMedCrossRef Utzschneider DT et al (2016) T cell factor 1-expressing memory-like CD8(+) T cells sustain the immune response to chronic viral infections. Immunity 45(2):415–427PubMedCrossRef
94.
go back to reference Bleakley M, Turtle CJ, Riddell SR (2012) Augmentation of anti-tumor immunity by adoptive T-cell transfer after allogeneic hematopoietic stem cell transplantation. Expert Rev Hematol 5(4):409–425PubMedPubMedCentralCrossRef Bleakley M, Turtle CJ, Riddell SR (2012) Augmentation of anti-tumor immunity by adoptive T-cell transfer after allogeneic hematopoietic stem cell transplantation. Expert Rev Hematol 5(4):409–425PubMedPubMedCentralCrossRef
95.
go back to reference Breems DA, Lowenberg B (2005) Autologous stem cell transplantation in the treatment of adults with acute myeloid leukaemia. Br J Haematol 130(6):825–833PubMedCrossRef Breems DA, Lowenberg B (2005) Autologous stem cell transplantation in the treatment of adults with acute myeloid leukaemia. Br J Haematol 130(6):825–833PubMedCrossRef
96.
go back to reference Guinan EC et al (1999) Transplantation of anergic histoincompatible bone marrow allografts. N Engl J Med 340(22):1704–1714PubMedCrossRef Guinan EC et al (1999) Transplantation of anergic histoincompatible bone marrow allografts. N Engl J Med 340(22):1704–1714PubMedCrossRef
97.
go back to reference Pikovskaya O et al (2016) Cutting edge: eomesodermin is sufficient to direct Type 1 innate lymphocyte development into the conventional NK lineage. J Immunol 196(4):1449–1454PubMedCrossRef Pikovskaya O et al (2016) Cutting edge: eomesodermin is sufficient to direct Type 1 innate lymphocyte development into the conventional NK lineage. J Immunol 196(4):1449–1454PubMedCrossRef
100.
go back to reference Goverman J, Perchellet A, Huseby ES (2005) The role of CD8(+) T cells in multiple sclerosis and its animal models. Curr Drug Targets Inflamm Allergy 4(2):239–245PubMedCrossRef Goverman J, Perchellet A, Huseby ES (2005) The role of CD8(+) T cells in multiple sclerosis and its animal models. Curr Drug Targets Inflamm Allergy 4(2):239–245PubMedCrossRef
101.
go back to reference Antia R, Ganusov VV, Ahmed R (2005) The role of models in understanding CD8+ T-cell memory. Nat Rev Immunol 5(2):101–111PubMedCrossRef Antia R, Ganusov VV, Ahmed R (2005) The role of models in understanding CD8+ T-cell memory. Nat Rev Immunol 5(2):101–111PubMedCrossRef
105.
go back to reference McQueen B et al (2016) Natural killer group 2D and CD28 receptors differentially activate mammalian/mechanistic target of rapamycin to alter murine effector CD8+ T-cell differentiation. Immunology 147(3):305–320PubMedPubMedCentralCrossRef McQueen B et al (2016) Natural killer group 2D and CD28 receptors differentially activate mammalian/mechanistic target of rapamycin to alter murine effector CD8+ T-cell differentiation. Immunology 147(3):305–320PubMedPubMedCentralCrossRef
Metadata
Title
TCF-1 regulates NKG2D expression on CD8 T cells during anti-tumor responses
Authors
Rebecca Harris
Mahinbanu Mammadli
Shannon Hiner
Liye Suo
Qi Yang
Jyoti Misra Sen
Mobin Karimi
Publication date
23-12-2022
Publisher
Springer Berlin Heidelberg
Published in
Cancer Immunology, Immunotherapy / Issue 6/2023
Print ISSN: 0340-7004
Electronic ISSN: 1432-0851
DOI
https://doi.org/10.1007/s00262-022-03323-0

Other articles of this Issue 6/2023

Cancer Immunology, Immunotherapy 6/2023 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine