Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Nat Immunol ; 23(3): 399-410, 2022 03.
Article in English | MEDLINE | ID: mdl-35145298

ABSTRACT

Targeted blockade of the checkpoint molecule programmed cell death 1 (PD-1) can activate tumor-specific T cells to destroy tumors, whereas targeted potentiation of PD-1 is expected to suppress autoreactive T cells and alleviate autoimmune diseases. However, the development of methods to potentiate PD-1 remains challenging. Here we succeeded in eliciting PD-1 function by targeting the cis-PD-L1-CD80 duplex, formed by binding of CD80 to the PD-1 ligand PD-L1, that attenuates PD-L1-PD-1 binding and abrogates PD-1 function. By generating anti-CD80 antibodies that detach CD80 from the cis-PD-L1-CD80 duplex and enable PD-L1 to engage PD-1 in the presence of CD80, we demonstrate that the targeted dissociation of cis-PD-L1-CD80 duplex elicits PD-1 function in the condition where PD-1 function is otherwise restricted. We demonstrate using murine models that the removal of PD-1 restriction is effective in alleviating autoimmune disease symptoms. Our findings establish a method to potentiate PD-1 function and propose the removal of restraining mechanisms as an efficient strategy to potentiate the function of inhibitory molecules.


Subject(s)
Autoimmune Diseases , Neoplasms , Animals , Autoimmunity , B7-1 Antigen , B7-H1 Antigen/metabolism , Mice , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes
2.
Immunity ; 55(5): 912-924.e8, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35413245

ABSTRACT

Lymphocyte activation gene-3 (LAG-3) is a potent inhibitory co-receptor; yet, its functional ligand remains elusive, with distinct potential ligands identified. Here, we investigated the relative contribution of potential ligands, stable peptide-MHC class II complexes (pMHCII) and fibrinogen-like protein 1 (FGL1), to LAG-3 activity in vitro and in vivo. Binding of LAG-3 to stable pMHCII but not to FGL1 induced T cell suppression in vitro. Consistently, LAG-3 mutants lacking FGL1-binding capacity but not those lacking stable pMHCII-binding capacity retained suppressive activity in vitro. Accordingly, targeted disruption of stable pMHCII- but not FGL1-binding capacity of LAG-3 in NOD mice recapitulated diabetes exacerbation by LAG-3 deficiency. Additionally, the loss of stable pMHCII-binding capacity of LAG-3 augmented anti-cancer immunity comparably with LAG-3 deficiency in C57BL/6 mice. These results identify stable pMHCII as a functional ligand of LAG-3 both in autoimmunity and anti-cancer immunity. Thus, stable pMHCII-LAG-3 interaction is a potential therapeutic target in human diseases.


Subject(s)
Antigens, CD , Autoimmunity , Histocompatibility Antigens Class II , Neoplasms , T-Lymphocytes , Animals , Antigens, CD/metabolism , Histocompatibility Antigens Class II/metabolism , Ligands , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Neoplasms/immunology , Peptides/metabolism , T-Lymphocytes/immunology , Lymphocyte Activation Gene 3 Protein
3.
Nat Immunol ; 19(12): 1415-1426, 2018 12.
Article in English | MEDLINE | ID: mdl-30349037

ABSTRACT

The success of tumor immunotherapy targeting the inhibitory co-receptors PD-1 and CTLA-4 has indicated that many other co-receptors might be potential druggable targets, despite limited information about their functional differences. Here we identified a unique target selectivity for the inhibitory co-receptor LAG-3 that was intrinsic to its immunoregulatory roles. Although LAG-3 has been reported to recognize major histocompatibility complex (MHC) class II, it did not recognize MHC class II universally; instead, we found that it selectively recognized stable complexes of peptide and MHC class II (pMHCII). LAG-3 did not directly interfere with interactions between the co-receptor CD4 and MHC class II or between the T cell antigen receptor and MHC class II. Instead, LAG-3 preferentially suppressed T cells responsive to stable pMHCII by transducing inhibitory signals via its intracellular region. Thus, LAG-3 might function more selectively than previously thought and thereby maintain tolerance to dominant autoantigens.


Subject(s)
Antigens, CD/immunology , CD4-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class II/immunology , Lymphocyte Activation/immunology , Animals , Antigens, CD/chemistry , Cell Line, Tumor , Humans , Mice , Molecular Conformation , Lymphocyte Activation Gene 3 Protein
4.
Mol Cell ; 77(5): 937-950.e6, 2020 03 05.
Article in English | MEDLINE | ID: mdl-31926851

ABSTRACT

Targeted blockade of programmed cell death 1 (PD-1), an immune-checkpoint receptor that inhibits T cell activation, provides clinical benefits in various cancers. However, how PD-1 modulates gene expression in T cells remains enigmatic. Here we investigated how PD-1 affects transcriptome changes induced by T cell receptor (TCR) activation. Intriguingly, we identified a huge variance in PD-1 sensitivity among TCR-inducible genes. When we quantified the half maximal effective concentration (EC50) as the relationship between change in gene expression and TCR signal strength, we found that genes associated with survival and proliferation were efficiently expressed upon TCR activation and resistant to PD-1-mediated inhibition. Conversely, genes encoding cytokines and effector molecules were expressed less efficiently and sensitive to PD-1-mediated inhibition. We further demonstrated that transcription factor binding motifs and CpG frequency in the promoter region affect EC50 and thus the PD-1 sensitivity of genes. Our findings explain how PD-1, dependent on the TCR signal strength, calibrates cellular transcriptomes to shape functional properties of T cell populations.


Subject(s)
Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/metabolism , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/metabolism , Transcriptome , Animals , Apoptosis , Binding Sites , Cell Proliferation , Coculture Techniques , CpG Islands , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation, Neoplastic , Genes, T-Cell Receptor , HEK293 Cells , Humans , Jurkat Cells , Lymphocytes, Tumor-Infiltrating/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Programmed Cell Death 1 Receptor/deficiency , Programmed Cell Death 1 Receptor/genetics , Promoter Regions, Genetic , Signal Transduction , T-Lymphocytes/immunology , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation
5.
Proc Natl Acad Sci U S A ; 120(1): e2210283120, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36577074

ABSTRACT

Single-cell whole-transcriptome analysis is the gold standard approach to identifying molecularly defined cell phenotypes. However, this approach cannot be used for dynamics measurements such as live-cell imaging. Here, we developed a multifunctional robot, the automated live imaging and cell picking system (ALPS) and used it to perform single-cell RNA sequencing for microscopically observed cells with multiple imaging modes. Using robotically obtained data that linked cell images and the whole transcriptome, we successfully predicted transcriptome-defined cell phenotypes in a noninvasive manner using cell image-based deep learning. This noninvasive approach opens a window to determine the live-cell whole transcriptome in real time. Moreover, this work, which is based on a data-driven approach, is a proof of concept for determining the transcriptome-defined phenotypes (i.e., not relying on specific genes) of any cell from cell images using a model trained on linked datasets.


Subject(s)
Deep Learning , Robotic Surgical Procedures , Robotics , Transcriptome , Image Processing, Computer-Assisted/methods , Gene Expression Profiling , Phenotype
6.
Nat Immunol ; 14(12): 1212-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24240160

ABSTRACT

PD-1, a negative coreceptor expressed on antigen-stimulated T cells and B cells, seems to serve as a 'rheostat' of the immune response. The molecular mechanisms of the functions of PD-1, in conjunction with the mild, chronic and strain-specific autoimmune phenotypes of PD-1-deficient mice, in contrast to the devastating fatal autoimmune disease of mice deficient in the immunomodulatory receptor CTLA-4, suggest that immunoregulation by PD-1 is rather antigen specific and is mainly cell intrinsic. Such unique properties make PD-1 a powerful target for immunological therapy, with highly effective clinical applications for cancer treatment.


Subject(s)
B-Lymphocytes/immunology , Immune System/immunology , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/immunology , Animals , B-Lymphocytes/metabolism , Humans , Immune System/metabolism , Immune Tolerance/immunology , Mice , Models, Immunological , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction/immunology , T-Lymphocytes/metabolism
7.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Article in English | MEDLINE | ID: mdl-34433672

ABSTRACT

Anti-PD-1 therapies can activate tumor-specific T cells to destroy tumors. However, whether and how T cells with different antigen specificity and affinity are differentially regulated by PD-1 remain vaguely understood. Upon antigen stimulation, a variety of genes is induced in T cells. Recently, we found that T cell receptor (TCR) signal strength required for the induction of genes varies across different genes and PD-1 preferentially inhibits the induction of genes that require stronger TCR signal. As each T cell has its own response characteristics, inducibility of genes likely differs across different T cells. Accordingly, the inhibitory effects of PD-1 are also expected to differ across different T cells. In the current study, we investigated whether and how factors that modulate T cell responsiveness to antigenic stimuli influence PD-1 function. By analyzing TCRs with different affinities to peptide-MHC complexes (pMHC) and pMHCs with different affinities to TCR, we demonstrated that PD-1 inhibits the expression of TCR-inducible genes efficiently when TCR:pMHC affinity is low. In contrast, affinities of peptides to MHC and MHC expression levels did not affect PD-1 sensitivity of TCR-inducible genes although they markedly altered the dose responsiveness of T cells by changing the efficiency of pMHC formation, suggesting that the strength of individual TCR signal is the key determinant of PD-1 sensitivity. Accordingly, we observed a preferential expansion of T cells with low-affinity to tumor-antigen in PD-1-deficient mice upon inoculation of tumor cells. These results demonstrate that PD-1 imposes qualitative control of T cell responses by preferentially suppressing low-affinity T cells.


Subject(s)
Antigens, Neoplasm/immunology , Lymphocyte Activation/immunology , Programmed Cell Death 1 Receptor/physiology , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Thymoma/immunology , Thymus Neoplasms/immunology , Animals , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , Receptors, Antigen, T-Cell/metabolism , Thymoma/metabolism , Thymoma/pathology , Thymus Neoplasms/metabolism , Thymus Neoplasms/pathology
8.
Int Immunol ; 33(12): 693-698, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34596210

ABSTRACT

Cancer immunotherapies that target PD-1 (programmed cell death 1) aim to destroy tumors by activating tumor-specific T cells that are otherwise inactivated by PD-1. Although these therapies have significantly improved the outcomes of patients with diverse cancer types and have revolutionized cancer treatment, only a limited proportion of patients benefits from the therapies currently. Therefore, there is a continued need to decipher the complex biology of PD-1 to improve therapeutic efficacies as well as to prevent immune-related adverse events. Especially, the spaciotemporal context in which PD-1 functions and the properties of T cells that are restrained by PD-1 are only vaguely understood. We have recently revealed that PD-1 function is strictly restricted at the activation phase of T-cell responses by the cis-interactions of PD-L1 and CD80 on antigen-presenting cells, which is critical for the induction of optimal T-cell responses. We also found that the sensitivity to the effects of PD-1 in T cells is essentially determined by T-cell-intrinsic factors. In T cells bearing T-cell antigen-receptors (TCRs) with lower affinity to antigenic peptides, PD-1 inhibits the expression of TCR-inducible genes more efficiently; thereby PD-1 preferentially suppresses low-affinity T cells. Thus, PD-1 function is coordinately regulated by various T-cell-intrinsic and -extrinsic factors that alter the responsiveness of T cells and the availability of PD-1 ligands. Precise and deeper understanding of the regulatory mechanisms of PD-1 is expected to facilitate the rational development of effective and safe immunotherapies.


Subject(s)
Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/immunology , Animals , Humans
9.
J Biol Chem ; 294(15): 6017-6026, 2019 04 12.
Article in English | MEDLINE | ID: mdl-30760527

ABSTRACT

T cell activation is tightly regulated by both stimulatory and inhibitory co-receptors and has been a focus in the development of interventions for managing cancer or autoimmune diseases. Targeting the inhibitory co-receptors programmed cell death 1 (PD-1) and cytotoxic T lymphocyte-associated protein 4 (CTLA-4) has successfully eradicated tumors but induced immune-related adverse events in humans and mice. The beneficial and adverse effects of targeting these co-receptors highlight their importance in cancer immunity and also autoimmunity. Although the therapeutic potencies of other inhibitory co-receptors are under extensive investigation, their inhibitory mechanisms and their functional differences are not well understood. Here we analyzed the inhibitory mechanisms of lymphocyte activation gene-3 (LAG-3), another inhibitory co-receptor, by using an in vitro T cell activation system and a high-affinity anti-LAG-3 antibody that strongly interferes with the binding of LAG-3 to its ligand. We found that the expression level of LAG-3 strongly correlates with the inhibitory function of LAG-3, suggesting that LAG-3 functions as a rheostat rather than as a breaker of T cell activation. By evaluating the inhibitory capacities of various LAG-3 variants relative to their expression levels, we found that LAG-3 transduces two independent inhibitory signals through an FXXL motif in the membrane-proximal region and the C-terminal EX repeat. These motifs have not been reported previously for inhibitory co-receptors, suggesting that LAG-3 inhibits T cell activation through a nonredundant inhibitory mechanisms along with the other inhibitory co-receptors. Our findings provide a rationale for combinatorial targeting of LAG-3 and the other inhibitory co-receptors to improve cancer immunotherapy.


Subject(s)
Antigens, CD/immunology , Gene Expression Regulation/immunology , Lymphocyte Activation , Signal Transduction/immunology , T-Lymphocytes/immunology , Amino Acid Motifs , Animals , Antigens, CD/genetics , Mice , Mice, Knockout , Protein Domains , Signal Transduction/genetics , Lymphocyte Activation Gene 3 Protein
10.
J Biol Chem ; 294(52): 19896-19906, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31723031

ABSTRACT

The inhibitory co-receptor programmed cell death 1 (PD-1, Pdcd1) plays critical roles in the regulation of autoimmunity, anticancer immunity, and immunity against infections. Immunotherapies targeting PD-1 have revolutionized cancer management and instigated various trials of improved cancer immunotherapies. Moreover, extensive trials are underway to potentiate PD-1 function to suppress harmful immune responses. Here we found that both natural and synthetic glucocorticoids (GCs) up-regulate PD-1 on T cells without altering the expression levels of other co-receptors and cell surface molecules. GC-induced up-regulation of PD-1 depended on transactivation of PD-1 transcription mediated through the glucocorticoid receptor. We further found that a GC response element 2525 bp upstream of the transcription start site of Pdcd1 is responsible for GC-mediated transactivation. We also observed that in vivo administration of GCs significantly up-regulates PD-1 expression on tumor-infiltrating T cells. By analyzing T cells differing in PD-1 expression, we directly demonstrated that the amount of PD-1 on the cell surface correlates with its inhibitory effect. Accordingly, GCs potentiated the capacity of PD-1 to inhibit T cell activation, suggesting that this PD-1-mediated inhibition contributes, at least in part, to the anti-inflammatory and immunosuppressive effects of GCs. In light of the critical roles of PD-1 in the regulation of autoimmunity, we expect that the potentiation of PD-1 activity may offer a promising therapeutic strategy for managing inflammatory and autoimmune diseases. Our current findings provide a rationale for strategies seeking to enhance the inhibitory effect of PD-1 by increasing its expression level.


Subject(s)
Glucocorticoids/pharmacology , Programmed Cell Death 1 Receptor/metabolism , Up-Regulation/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Dexamethasone/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/genetics , Promoter Regions, Genetic , Receptors, Glucocorticoid/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transcription Initiation Site , Transcriptional Activation/drug effects
11.
Biochem Biophys Res Commun ; 511(3): 491-497, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30771904

ABSTRACT

Cancer immunotherapies targeting programmed cell death 1 (PD-1) and cytotoxic T-lymphocyte antigen 4 revolutionized cancer treatment and instigated various trials to develop new cancer immunotherapies with higher therapeutic efficacy. Agonistic Abs against tumor necrosis factor receptor super family (TNFRSF) molecules are highly expected due to their high potential to enhance survival, proliferation, and effector function of T cells. To date, agonistic antibodies (Abs) against CD27, GITR, OX40, and 4-1BB have been reported to increase the efficacy of anti-PD-1 therapy in animal models and clinical trials of these combinatorial therapies are underway. However, the mechanisms how agonistic Abs against TNFRSF molecules potentiate anti-PD-1 therapy are not well understood. Here we examined the potency of PD-1 to inhibit the antigen-dependent activation of T cells in the presence of co-stimulation through CD27 and GITR by using in vitro and ex vivo co-culture systems of T cells and antigen presenting cells. The cytokine secretion from T cells upon antigen stimulation was strongly augmented by the engagement of CD27 or GITR with their corresponding ligands. Remarkably, PD-1 efficiently inhibited the activation of T cells even in the presence of co-stimulation through CD27 or GITR. Accordingly, cytokine secretion was synergistically augmented when PD-1 blockade was combined with triggering of CD27 or GITR. These results indicate that the triggering of TNFRSF molecules and PD-1 blockade can act on the same individual cells simultaneously to augment the magnitude of T cell activation, providing the rationale for the combinatorial usage of agonistic Abs against TNFRSF molecules and blocking Abs against PD-1 or PD-L1.


Subject(s)
Glucocorticoid-Induced TNFR-Related Protein/immunology , Lymphocyte Activation , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Animals , Cell Line, Tumor , Cells, Cultured , Mice, Inbred BALB C
12.
J Autoimmun ; 105: 102296, 2019 12.
Article in English | MEDLINE | ID: mdl-31277964

ABSTRACT

Anti-PD-1 therapy can induce eradication of tumors and immune-related adverse events (irAEs) in humans and model animals. However, how anti-PD-1 therapy modifies cellular phenotypes of CD8+ T cells to destroy tumors and damage self-tissues remains to be clarified. Here we performed single cell mRNA expression profiling of autoreactive CD8+ T cells under or beyond PD-1 suppression in target tissues and reconstructed their activation trajectory. Autoreactive CD8+ T cells went through four activation phases and PD-1 strongly attenuated the transition from the second- to the third-phase, where effector functions were acquired. Shifts in cluster composition of autoreactive CD8+ T cells markedly reflected the severity of autoimmunity. In addition, genes up-regulated along the activation-trajectory in autoimmunity were highly expressed in responders of melanoma patients in anti-PD-1 therapy, suggesting that tumor-specific T cells need to be activated in a similar trajectory to destroy tumors in human patients upon PD-1 blockade. These findings reveal that PD-1 blockade facilitates the activation trajectory of CD8+ T cells to boost their effector functions. Targeted manipulation of the trajectory could lead to new therapeutic opportunities.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocyte Activation/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , Antibodies, Monoclonal/immunology , Autoimmunity/immunology , Cell Line, Tumor , Female , Humans , Melanoma/immunology , Mice , Mice, Inbred NOD
13.
Adv Exp Med Biol ; 1189: 213-232, 2019.
Article in English | MEDLINE | ID: mdl-31758536

ABSTRACT

Co-receptors cooperatively regulate the function of immune cells to optimize anti-infectious immunity while limiting autoimmunity by providing stimulatory and inhibitory co-signals. Among various co-receptors, those in the CD28/CTLA-4 family play fundamental roles in the regulation of lymphocytes by modulating the strength, quality, and/or duration of the antigen receptor signal. The development of the lethal lymphoproliferative disorder and various tissue-specific autoimmune diseases in mice deficient for CTLA-4 and PD-1, respectively, clearly demonstrates their pivotal roles in the development and the maintenance of immune tolerance. The recent success of immunotherapies targeting CTLA-4 and PD-1 in the treatment of various cancers highlights their critical roles in the regulation of cancer immunity in human. In addition, the development of multifarious autoimmune diseases as immune-related adverse events of anti-CTLA-4 and anti-PD-1/PD-L1 therapies and the successful clinical application of the CD28 blocking therapy using CTLA-4-Ig to the treatment of arthritis assure their crucial roles in the regulation of autoimmunity in human. Accumulating evidences in mice and humans indicate that genetic and environmental factors strikingly modify effects of the targeted inhibition and potentiation of co-signals. In this review, we summarize our current understanding of the roles of CD28, CTLA-4, and PD-1 in autoimmunity. Deeper understandings of the context-dependent and context-independent functions of co-signals are essential for the appropriate usage and the future development of innovative immunomodulatory therapies for a diverse array of diseases.


Subject(s)
Autoimmunity , CD28 Antigens/metabolism , CTLA-4 Antigen/metabolism , Programmed Cell Death 1 Receptor/metabolism , Animals , Humans , Immune Tolerance , Immunotherapy , Mice
14.
J Autoimmun ; 86: 75-92, 2018 01.
Article in English | MEDLINE | ID: mdl-28931462

ABSTRACT

Autoimmunity is prevented by the function of the autoimmune regulator [AIRE (Aire in mice)], which promotes the expression of a wide variety of tissue-restricted antigens (TRAs) from medullary thymic epithelial cells (mTECs) and from a subset of peripheral antigen-presenting cells (APCs). We examined the effect of additive expression of human AIRE (huAIRE) in a model of autoimmune diabetes in NOD mice. Unexpectedly, we observed that mice expressing augmented AIRE/Aire developed muscle-specific autoimmunity associated with incomplete maturation of mTECs together with impaired expression of Aire-dependent TRAs. This led to failure of deletion of autoreactive T cells together with dramatically reduced production of regulatory T cells in the thymus. In peripheral APCs, expression of costimulatory molecules was augmented. We suggest that levels of Aire expression need to be tightly controlled for maintenance of immunological tolerance. Our results also highlight the importance of coordinated action between central tolerance and peripheral tolerance under the common control of Aire.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Muscles/immunology , Polymyositis/immunology , Thymus Gland/immunology , Transcription Factors/metabolism , Animals , Autoantigens/metabolism , Autoimmunity , Disease Models, Animal , Humans , Immune Tolerance , Mice , Mice, Inbred NOD , Mice, Transgenic , Organ Specificity , Transcription Factors/genetics , AIRE Protein
15.
Cancer Sci ; 107(2): 189-202, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26919617

ABSTRACT

Non-clinical studies are necessary at each stage of the development of oncology drugs. Many experimental cancer models have been developed to investigate carcinogenesis, cancer progression, metastasis, and other aspects in cancer biology and these models turned out to be useful in the efficacy evaluation and the safety prediction of oncology drugs. While the diversity and the degree of engagement in genetic changes in the initiation of cancer cell growth and progression are widely accepted, it has become increasingly clear that the roles of host cells, tissue microenvironment, and the immune system also play important roles in cancer. Therefore, the methods used to develop oncology drugs should continuously be revised based on the advances in our understanding of cancer. In this review, we extensively summarize the effective use of those models, their advantages and disadvantages, ranges to be evaluated and limitations of the models currently used for the development and for the evaluation of oncology drugs.


Subject(s)
Disease Models, Animal , Drug Discovery/methods , Medical Oncology/methods , Neoplasms , Animals , Antineoplastic Agents , Humans
16.
Proc Natl Acad Sci U S A ; 109(13): 5010-5, 2012 Mar 27.
Article in English | MEDLINE | ID: mdl-22403066

ABSTRACT

Adequate activation of CD4(+) T lymphocytes is essential for host defense against invading pathogens; however, exaggerated activity of effector CD4(+) T cells induces tissue damage, leading to inflammatory disorders such as inflammatory bowel diseases. Several unique subsets of intestinal innate immune cells have been identified. However, the direct involvement of innate immune cell subsets in the suppression of T-cell-dependent intestinal inflammation is poorly understood. Here, we report that intestinal CX(3)C chemokine receptor 1(high) (CX(3)CR1(high)) CD11b(+) CD11c(+) cells are responsible for prevention of intestinal inflammation through inhibition of T-cell responses. These cells inhibit CD4(+) T-cell proliferation in a cell contact-dependent manner and prevent T-cell-dependent colitis. The suppressive activity is abrogated in the absence of the IL-10/Stat3 pathway. These cells inhibit T-cell proliferation by two steps. Initially, CX(3)CR1(high) CD11b(+) CD11c(+) cells preferentially interact with T cells through highly expressed intercellular adhesion molecule-1/vascular cell adhesion molecule-1; then, they fail to activate T cells because of defective expression of CD80/CD86. The IL-10/Stat3 pathway mediates the reduction of CD80/CD86 expression. Transfer of wild-type CX(3)CR1(high) CD11b(+) CD11c(+) cells prevents development of colitis in myeloid-specific Stat3-deficient mice. Thus, these cells are regulatory myeloid cells that are responsible for maintaining intestinal homeostasis.


Subject(s)
Colitis/immunology , Colitis/pathology , Intestines/immunology , Intestines/pathology , Myeloid Cells/immunology , Receptors, Chemokine/metabolism , T-Lymphocytes/immunology , Animals , CD11 Antigens/metabolism , CX3C Chemokine Receptor 1 , Cell Proliferation , Colitis/complications , Colitis/prevention & control , Inflammation/complications , Inflammation/immunology , Inflammation/pathology , Inflammation/prevention & control , Interleukin-10/metabolism , Mice , Mucous Membrane/immunology , Mucous Membrane/pathology , STAT3 Transcription Factor/deficiency , STAT3 Transcription Factor/metabolism
17.
Clin Immunol ; 146(1): 15-25, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23178752

ABSTRACT

It is unclear what roles TNF-α has in the development of autoimmune hepatitis (AIH) and whether AIH is responsive to anti-TNF-α. We recently developed a mouse model of fatal AIH that develops in PD-1-deficient mice thymectomized three days after birth, finding that CCR6-CCL20 axis-dependent migration of dysregulated splenic T cells is crucial to induce AIH. In this study, we show the indispensable role of TNF-α in the development of AIH. Administering anti-TNF-α prevented the induction, but treatment by anti-TNF-α after the induction did not suppress progression. Administering anti-TNF-α did not prevent splenic T-cell activation, but did suppress hepatic CCL20 expression. In contrast, administering anti-CCL20 suppressed AIH but not elevated serum TNF-α levels. TNF-α stimulation enhanced CCL20 expression in hepatocytes. These findings suggest that TNF-α is essential in the induction of AIH through upregulation of hepatic CCL20 expression, which allows migration of dysregulated splenic T cells.


Subject(s)
Chemokine CCL20/immunology , Hepatitis, Autoimmune/immunology , Liver/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Cell Movement/drug effects , Cell Movement/immunology , Cells, Cultured , Chemokine CCL20/genetics , Chemokine CCL20/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression/drug effects , Hepatitis, Autoimmune/mortality , Hepatitis, Autoimmune/prevention & control , Hepatocytes/drug effects , Hepatocytes/immunology , Hepatocytes/metabolism , Kaplan-Meier Estimate , Liver/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Programmed Cell Death 1 Receptor/deficiency , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Reverse Transcriptase Polymerase Chain Reaction , Spleen/immunology , Spleen/metabolism , Survival Rate , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymectomy , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects
18.
J Immunol ; 186(5): 2772-9, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21263073

ABSTRACT

Programmed cell death-1 (PD-1) is an inhibitory coreceptor for T lymphocytes that provides feedback inhibition of T cell activation. Although PD-1's expression on T cells is known to be activation dependent, the factors that determine the timing, intensity, and duration of PD-1 expression in immune reactions are not fully understood. To address this question, we performed a fine mapping analysis of a conserved 5'-flanking region of the PD-1 gene and identified a putative IFN stimulation response element, which was responsible for PD-1 transcription in the 2B4.11 T cell line. Consistent with this finding, activation by IFN-α enhanced both the induction and maintenance of PD-1 expression on TCR-engaged primary mouse T cells through an association IFN-responsive factor 9 (IRF9) to the IFN stimulation response element. Furthermore, PD-1 expression on Ag-specific CD8(+) T cells was augmented by IFN-α in vivo. We propose that strong innate inflammatory responses promote primary T cell activation and their differentiation into effector cells, but also cause an attenuated T cell response in sustained immune reactions, at least partially through type I IFN-mediated PD-1 transcription. Based on this idea, we demonstrate that IFN-α administration in combination with PD-1 blockade in tumor-bearing mice effectively augments the antitumor immunity, and we propose this as a novel and rational approach for cancer immunotherapy.


Subject(s)
Antigens, Surface/genetics , Apoptosis Regulatory Proteins/genetics , Immunity, Cellular , Interferon-alpha/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transcription, Genetic/immunology , Animals , Antigens, Surface/biosynthesis , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/biosynthesis , Cell Line , Cell Line, Tumor , Cells, Cultured , Humans , Immunity, Cellular/genetics , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Interferon-Stimulated Gene Factor 3, gamma Subunit/physiology , Interferon-alpha/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Programmed Cell Death 1 Receptor , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/physiology , Regulatory Elements, Transcriptional/immunology , Signal Transduction/genetics , Signal Transduction/immunology
19.
Gastroenterology ; 140(4): 1322-1333.e1-5, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21237169

ABSTRACT

BACKGROUND & AIMS: To clarify mechanisms involved in the development of autoimmune hepatitis (AIH), we recently developed a mouse model of spontaneous AIH by inducing a concurrent loss of Foxp3(+) regulatory T cells and programmed cell death 1 (PD-1)-mediated signaling. Fatal AIH in these mice was characterized by severe T-cell infiltration and huge production of antinuclear antibodies (Abs). This study aims to identify induction sites, responsible T-cell subsets, and key molecules for induction of AIH. METHODS: To develop the mouse model of AIH, neonatal thymectomy (NTx) was performed on PD-1-deficient (PD-1(-/-)) mice. We then conducted neonatal splenectomy or in vivo administration of Abs to cytokines, chemokines, or cell-surface molecules. RESULTS: In NTx-PD-1(-/-) mice, either neonatal splenectomy or in vivo CD4(+) T-cell depletion suppressed CD4(+) and CD8(+) T-cell infiltration in the liver. In the induction phase of AIH, splenic CD4(+) T cells were localized in B-cell follicles with huge germinal centers and showed the Bcl6(+) inducible costimulator (ICOS)(+) interleukin (IL)-21(+) IL-21 receptor (IL-21R)(+) follicular helper T (T(FH)) cell phenotype. Blocking Abs to ICOS or IL-21 suppressed T(FH)-cell generation and induction of AIH. In addition, IL-21 produced by T(FH) cells drove CD8(+) T-cell activation. Splenic T(FH) cells and CD8(+) T cells expressed CCR6, and CCL20 expression was elevated in the liver. Administration of anti-CCL20 suppressed migration of these T cells to the liver and induction of AIH. CONCLUSIONS: Dysregulated T(FH) cells in the spleen are responsible for the induction of fatal AIH, and CCR6-CCL20 axis-dependent migration of splenic T cells is crucial to induce AIH in NTx-PD-1(-/-) mice.


Subject(s)
Hepatitis, Autoimmune/immunology , Hepatitis, Autoimmune/pathology , Spleen/immunology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/pathology , Animals , Antigens, Differentiation, T-Lymphocyte/immunology , Antigens, Differentiation, T-Lymphocyte/metabolism , Antigens, Surface/genetics , Antigens, Surface/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Movement/immunology , Chemokine CCL20/immunology , Chemokine CCL20/metabolism , Disease Models, Animal , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Germinal Center/cytology , Germinal Center/immunology , Hepatitis, Autoimmune/mortality , Inducible T-Cell Co-Stimulator Protein , Interleukins/immunology , Interleukins/metabolism , Liver/immunology , Liver/pathology , Mice , Mice, Mutant Strains , Programmed Cell Death 1 Receptor , Receptors, CCR6/immunology , Receptors, CCR6/metabolism , Signal Transduction/immunology , Spleen/cytology , Spleen/surgery , Splenectomy , Thymectomy
20.
Curr Top Microbiol Immunol ; 350: 17-37, 2011.
Article in English | MEDLINE | ID: mdl-21061197

ABSTRACT

Programmed cell death-1 (PD-1) is a member of the CD28 superfamily that delivers negative signals upon interaction with its two ligands, PD-L1 or PD-L2. PD-1 and its ligands are broadly expressed and exert a wider range of immunoregulatory roles in T cells activation and tolerance compared with other CD28 members. Subsequent studies show that PD-1-PD-L interaction regulates the induction and maintenance of peripheral tolerance and protect tissues from autoimmune attack. PD-1 and its ligands are also involved in attenuating infectious immunity and tumor immunity, and facilitating chronic infection and tumor progression. The biological significance of PD-1 and its ligand suggests the therapeutic potential of manipulation of PD-1 pathway against various human diseases. In this review, we summarize our current understanding of PD-1 and its ligands ranging from discovery to clinical significance.


Subject(s)
Antigens, CD/metabolism , Apoptosis Regulatory Proteins/metabolism , Autoimmune Diseases/immunology , Gene Expression Regulation/immunology , Neoplasms/immunology , T-Lymphocytes/immunology , Virus Diseases/immunology , Animals , Antibodies, Monoclonal/therapeutic use , Antigens, CD/chemistry , Antigens, CD/genetics , Antigens, CD/immunology , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/immunology , Chronic Disease , Clinical Trials, Phase I as Topic , Humans , Ligands , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Molecular , Neoplasms/therapy , Programmed Cell Death 1 Receptor , Treatment Outcome , Virus Diseases/therapy
SELECTION OF CITATIONS
SEARCH DETAIL