Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
Int J Mol Sci ; 22(4)2021 Feb 16.
Article in English | MEDLINE | ID: mdl-33669419

ABSTRACT

We have established an immune cell therapy with immortalized induced pluripotent stem-cell-derived myeloid lines (iPS-ML). The benefits of using iPS-ML are the infinite proliferative capacity and ease of genetic modification. In this study, we introduced 4-1BBL gene to iPS-ML (iPS-ML-41BBL). The analysis of the cell-surface molecules showed that the expression of CD86 was upregulated in iPS-ML-41BBL more than that in control iPS-ML. Cytokine array analysis was performed using supernatants of the spleen cells that were cocultured with iPS-ML or iPS-ML-41BBL. Multiple cytokines that are beneficial to cancer immunotherapy were upregulated. Peritoneal injections of iPS-ML-41BBL inhibited tumor growth of peritoneally disseminated mouse melanoma and prolonged survival of mice compared to that of iPS-ML. Furthermore, the numbers of antigen-specific CD8+ T cells were significantly increased in the spleen and tumor tissues treated with epitope peptide-pulsed iPS-ML-41BBL compared to those treated with control iPS-ML. The number of CXCR6-positive T cells were increased in the tumor tissues after treatment with iPS-ML-41BBL compared to that with control iPS-ML. These results suggest that iPS-ML-41BBL could activate antigen-specific T cells and promote their infiltration into the tumor tissues. Thus, iPS-ML-41BBL may be a candidate for future immune cell therapy aiming to change immunological "cold tumor" to "hot tumor".


Subject(s)
4-1BB Ligand/metabolism , CD8-Positive T-Lymphocytes/immunology , Immunotherapy/methods , Induced Pluripotent Stem Cells/cytology , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/therapy , Myeloid Cells/metabolism , Myeloid Cells/transplantation , Skin Neoplasms/therapy , Animals , Cell Line, Tumor , Cytokines/metabolism , Disease Models, Animal , Female , Melanoma/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, CXCR6/metabolism , Skin Neoplasms/pathology , Treatment Outcome
2.
Cancer Sci ; 110(10): 3027-3037, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31348591

ABSTRACT

We previously established a method to generate myeloid cells with a proliferative capability from pluripotent stem cells and designated them iPS-ML. Human iPS-ML cells share features with physiological macrophages including the capability to infiltrate into cancer tissues. We observed therapeutic effects of human iPS-ML cells expressing interferon ß (iPS-ML/interferon (IFN)-ß) in xenograft cancer models. However, assessment of host immune system-mediated therapeutic and adverse effects of this therapy is impossible by xenograft models. We currently evaluated the therapeutic effects of a mouse equivalent of human iPS-ML/IFN, a mouse embryonic stem (ES) cell-derived myeloid cell line producing IFN (ES-ML/IFN). The ES-MLs producing IFN-ß (ß-ML) and IFN-γ (γ-ML) and originating from E14 ES cells derived from the 129 mouse strain (H-2b ) were generated, and the MHC (H-2Kb , Db , and I-Ab ) genes of the ES-ML/IFN were disrupted using the clustered regularly interspaced short palindromic repeats (CRISPR)/CAS9 method. We used the ES-ML/IFN to treat allogeneic BALB/c mice (H-2d ) transplanted with Colon26 cancer cells. Treatment with ß-ML but not with γ-ML cells repressed the growth of colon cancer in the peritoneal cavity and liver. The transferred ES-ML/IFN infiltrated into cancer tissues and enhanced infiltration of T cells into cancer tissues. ES-ML/IFN therapy increased the number of immune cells in the lymphoid organs. Sensitization of both cancer antigen-specific CD8+ T cells and natural killer (NK) cells were enhanced by the therapy, and CD8+ T cells were essential for the therapeutic effect, implying that donor MHC-deficient ß-ML exhibited a therapeutic effect through the activation of host immune cells derived from allogeneic recipient mice. The results suggested the usefulness of HLA-deficient human iPS-ML/IFN-ß cells for therapy of HLA-mismatched allogeneic cancer patients.


Subject(s)
Colonic Neoplasms/therapy , Embryonic Stem Cells/cytology , Histocompatibility Antigens/genetics , Interferon-beta/metabolism , Myeloid Cells/transplantation , Animals , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Colonic Neoplasms/immunology , Embryonic Stem Cells/metabolism , Female , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Killer Cells, Natural/metabolism , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Mice, Inbred BALB C , Myeloid Cells/cytology , Myeloid Cells/metabolism , Transplantation, Homologous , Xenograft Model Antitumor Assays
3.
Biochem Biophys Res Commun ; 511(3): 711-717, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30827508

ABSTRACT

Immunogenicity of immature pluripotent stem cells is a topic of intense debate. Immunogenic antigens, which are specific in pluripotent states, have not been described previously. In this study, we identified glypican-3 (GPC3), a known carcinoembryonic antigen, as a pluripotent state-specific immunogenic antigen. Additionally, we validated the applicability of human leukocyte antigen (HLA)-class I-restricted GPC3-reactive cytotoxic T lymphocytes (CTLs) in the removal of undifferentiated pluripotent stem cells (PSCs) from human induced pluripotent stem cell (hiPSC)-derivatives. HiPSCs uniquely express GPC3 in pluripotent states and were rejected by GPC3-reactive CTLs, which were sensitized with HLA-class I-restricted GPC3 peptides. Furthermore, GPC3-reactive CTLs selectively removed undifferentiated PSCs from hiPSC-derivatives in vitro and inhibited tumor formation in vivo. Our results demonstrate that GPC3 works as a pluripotent state-specific immunogenic antigen in hiPSCs and is applicable to regenerative medicine as a method of removing undifferentiated PSCs, which are the main cause of tumor formation.


Subject(s)
Glypicans/immunology , Induced Pluripotent Stem Cells/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Differentiation , Cell Line , Glypicans/analysis , HLA-A2 Antigen/immunology , Humans , Induced Pluripotent Stem Cells/cytology , Mice, Inbred NOD , Mice, SCID , Models, Molecular , Neoplasms/immunology
4.
Cancer Sci ; 109(3): 523-530, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29090850

ABSTRACT

Accompanied by the growing clinical applications of immunotherapy in the treatment of cancer patients, development of novel therapeutic approaches to reverse the immune-suppressive environment in cancer patients is eagerly anticipated, because the success of cancer immunotherapy is currently limited by immune-suppressive effects in tumor-bearing hosts. Interleukin (IL)-6, a pleotropic proinflammatory cytokine, participates in tumor cell-autonomous processes that are required for their survival and growth, and is therefore known as a poor prognostic factor in cancer patients. In addition, an emerging role of IL-6 in modulating multiple functions of immune cells including T cells, dendritic cells, and macrophages is responsible for the dysfunction of innate and adaptive immunity against tumors. Therefore, the IL-6-targeting approach is of value as a promising strategy for desensitization and prevention of immune-suppressive effects, and should be an effective treatment when combined with current immunotherapies. The aim of the present review is to discuss the immune-suppressive aspects of IL-6, notably with modification of T-cell functions in cancer patients, and their relationship to anti-tumor immune responses and cancer immunotherapy.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Interleukin-6/metabolism , Neoplasms/drug therapy , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Clinical Trials as Topic , Drug Therapy , Humans , Immunotherapy , Interleukin-6/antagonists & inhibitors , Neoplasms/immunology , Signal Transduction , T-Lymphocytes/drug effects
5.
Cancer Sci ; 109(11): 3403-3410, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30142694

ABSTRACT

Although first-line chemotherapy has a high rate of complete responses in ovarian cancer patients, the vast majority of patients present with recurrent disease that has become refractory to conventional chemotherapy. Peritoneal dissemination and malignant ascites are the hallmarks of recurrent or advanced ovarian cancer and severely reduce quality of life. Development of therapeutic measures to treat such patients is eagerly anticipated. Macrophage infiltration is observed in various types of cancer including epithelial ovarian cancer. In addition, macrophages are involved in the formation of spheroids in the malignant ascites of ovarian cancer and promote cancer growth. iPS-ML, macrophage-like myelomonocytic cells generated from human induced pluripotent stem (iPS) cells, made close contacts with ovarian cancer cells in vitro. We hypothesized that, if we inoculate iPS-ML-producing IFN-ß (iPS-ML/IFN-ß) into the peritoneal cavity of patients with ovarian cancer, IFN-ß produced by the iPS-ML/IFN-ß would efficiently act on the cancer cells to suppress cancer growth. To evaluate this hypothesis, we injected iPS-ML/IFN-ß into SCID mice bearing peritoneally disseminated human ovarian cancer cells, SKOV3. Immunohistochemical analysis of the intraperitoneal tumors detected iPS-ML/IFN-ß infiltrating into the cancer tissues. Therapy with iPS-ML/IFN-ß significantly suppressed tumor progression. In addition, dramatic reduction of cancer-related ascites was observed. Collectively, it is suggested that iPS-ML/IFN-ß therapy offers a new approach for the treatment of patients with advanced ovarian cancer.


Subject(s)
Ascites/therapy , Interferon-beta/metabolism , Monocytes/transplantation , Ovarian Neoplasms/therapy , Peritoneal Neoplasms/therapy , Animals , Ascites/etiology , Cell Line, Tumor , Coculture Techniques , Female , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/immunology , Mice , Mice, SCID , Monocytes/cytology , Monocytes/immunology , Ovarian Neoplasms/complications , Ovarian Neoplasms/immunology , Peritoneal Neoplasms/complications , Peritoneal Neoplasms/immunology , Treatment Outcome , Xenograft Model Antitumor Assays
6.
J Gen Virol ; 99(9): 1239-1247, 2018 09.
Article in English | MEDLINE | ID: mdl-30058991

ABSTRACT

The lack of an appropriate model has been a serious concern in dengue research pertinent to immune response and vaccine development. It remains a matter of impediment in dengue virus (DENV) studies when it comes to an in vitro model, which requires adequate quantity of dendritic cells (DC) with uniform characters. Other sources of DC, mostly monocyte derived DC (moDC), have been used despite their limitations such as quantity, proliferation, and donor dependent characters. Recent development of human iPS cells with consistent proliferation for long, stable, functional characteristics and desired HLA background has certainly offered added advantages. Therefore, we hypothesised that iPS derived cells would be a reliable alternative to the traditional DCs to be used with an in vitro DENV system. To develop a DENV infection and T cell activation model, we utilised iPS cells (HLA-A*24) as the source of DC. iPS-ML-DC was prepared and DENV infectivity was assessed apart from the major surface markers expression and cytokine production potential. Our iPS-ML-DC had major DC markers expression, DENV infection efficiency and cytokine production properties similar to that of moDC. Moreover, DENV infected iPS-ML-DC demonstrated the ability to activate HLA-matched T cell (but not mismatched) in vitro as evidenced by significantly higher proportion of IFN-γ+ CD69+ T cells compared to non-infected iPS-ML-DC. This affirmed the antigen-specific T cell activation by iPS-ML-DC as a function of antigen presenting cells. To conclude, maturation potential, DENV infection efficiency and T cell activation ability collectively suggest that iPS-ML-DC serves as an attractive option of DC for use in DENV studies in vitro.


Subject(s)
Dendritic Cells/physiology , Dendritic Cells/virology , Dengue Virus/physiology , Induced Pluripotent Stem Cells , Avian Proteins/genetics , Avian Proteins/metabolism , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation , Humans , Lymphocyte Activation , T-Lymphocytes/physiology , Virus Replication/physiology
7.
J Biol Chem ; 291(48): 25096-25105, 2016 Nov 25.
Article in English | MEDLINE | ID: mdl-27758856

ABSTRACT

Familial amyloidotic polyneuropathy (FAP) is a systemic amyloidosis mainly caused by amyloidogenic transthyretin (ATTR). This incurable disease causes death ∼10 years after onset. Although it has been widely accepted that conformational change of the monomeric form of transthyretin (TTR) is very important for amyloid formation and deposition in the organs, no effective therapy targeting this step is available. In this study, we generated a mouse monoclonal antibody, T24, that recognized the cryptic epitope of conformationally changed TTR. T24 inhibited TTR accumulation in FAP model rats, which expressed human ATTR V30M in various tissues and exhibited non-fibrillar deposits of ATTR in the gastrointestinal tracts. Additionally, humanized T24 (RT24) inhibited TTR fibrillation and promoted macrophage phagocytosis of aggregated TTR. This antibody did not recognize normal serum TTR functioning properly in the blood. These results demonstrate that RT24 would be an effective novel therapeutic antibody for FAP.


Subject(s)
Amyloid Neuropathies, Familial/drug therapy , Amyloid Neuropathies, Familial/immunology , Antibodies, Monoclonal, Murine-Derived/immunology , Macrophages/immunology , Phagocytosis/drug effects , Prealbumin/immunology , Amyloid Neuropathies, Familial/pathology , Animals , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Female , Humans , Macrophages/pathology , Male , Mice , Rats
8.
J Immunol ; 193(4): 2024-33, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25031460

ABSTRACT

We established a method to generate a large quantity of myeloid lineage cells from mouse embryonic stem (ES) cells, termed ES cell-derived proliferating myeloid cell lines (ES-ML). ES-ML continuously proliferated in the presence of M-CSF and GM-CSF. ES-ML genetically modified to express an anti-HER2 (neu) mAb single-chain V region fragment reduced the number of cocultured mouse Colon-26 cancer cells expressing HER2. Stimulation of ES-ML with IFN-γ plus LPS or TNF resulted in almost complete killing of the Colon-26 cells by the ES-ML, and the cytotoxicity was mediated, in part, by NO produced by ES-ML. When ES-ML were injected into mice with i.p. established Colon-26 tumors, they efficiently infiltrated the tumor tissues. Injection of ES-ML with rIFN-γ and LPS inhibited cancer progression in the mouse peritoneal cavity. Coinjection of TNF-transfected or untransfected ES-ML with rIFN-γ inhibited cancer growth and resulted in prolonged survival of the treated mice. In this experiment, transporter associated with Ag processing (TAP)1-deficient ES-ML exhibited therapeutic activity in MHC-mismatched allogeneic recipient mice. Despite the proliferative capacity of ES-ML, malignancy never developed from the transferred ES-ML in the recipient mice. In summary, TAP-deficient ES-ML with anticancer properties exhibited a therapeutic effect in allogeneic recipients, suggesting the possible use of TAP-deficient human-induced pluripotent stem cell-derived proliferating myeloid cell lines in cancer therapy.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Colonic Neoplasms/therapy , Embryonic Stem Cells/immunology , Macrophages/immunology , Animals , Antibodies, Monoclonal/immunology , Cell Differentiation/immunology , Cell Line, Tumor , Colonic Neoplasms/immunology , Cytotoxicity, Immunologic , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Interferon-gamma/pharmacology , Lipopolysaccharides , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pluripotent Stem Cells/immunology , Receptor, ErbB-2/immunology , Single-Chain Antibodies/immunology , Transplantation, Homologous
9.
Rinsho Ketsueki ; 57(8): 1074-9, 2016 08.
Article in Japanese | MEDLINE | ID: mdl-27599426

ABSTRACT

Antibody-based anti-cancer immunotherapy was recently recognized as one of the truly effective therapies for cancer patients. Antibodies against cell surface cancer antigens, such as CD20, and also those against immune-inhibitory molecules called "immune checkpoint blockers", such as CTLA4 or PD1, have emerged. Large-scale clinical trials have confirmed that, in some cases, antibody-based drugs are superior to conventional chemotherapeutic agents. These antibody-based drugs are now being manufactured employing a mass-production system by pharmaceutical companies. Anti-cancer therapy by immune cells, i.e. cell-based immunotherapy, is expected to be more effective than antibody therapy, because immune cells can recognize, infiltrate, and act in cancer tissues more directly than antibodies. In order to achieve cell-based anti-cancer immunotherapy, it is necessary to develop manufacturing systems for mass-production of immune cells. Our group has been studying immunotherapy with myeloid cells derived from ES cells or iPS cells. These pluripotent stem cells can be readily propagated under constant culture conditions, with expansion into a large quantity. We consider these stem cells to be the most suitable cellular source for mass-production of immune cells. This review introduces our studies on anti-cancer therapy with iPS cell-derived dendritic cells and iPS cell-derived macrophages.


Subject(s)
Dendritic Cells/immunology , Immunotherapy , Induced Pluripotent Stem Cells/cytology , Macrophages/immunology , Neoplasms/therapy , Animals , Cell Transformation, Neoplastic , Dendritic Cells/cytology , Humans , Macrophages/cytology , Neoplasms/immunology
10.
Int J Cancer ; 134(2): 352-66, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24734272

ABSTRACT

We recently identified a novel cancer-testis antigen, cell division cycle associated 1 (CDCA1) using genome-wide cDNA microarray analysis, and CDCA1-derived cytotoxic T lymphocyte (CTL)-epitopes. In this study, we attempted to identify CDCA1-derived long peptides (LPs) that induce both CD4+ helper T (Th) cells and CTLs. We combined information from a recently developed computer algorithm predicting HLA class II-binding peptides with CDCA1-derived CTL-epitope sequences presented by HLA-A2 (A*02:01) or HLA-A24 (A*24:02) to select candidate CDCA1-LPs encompassing both Th cell epitopes and CTL-epitopes. We studied the immunogenicity of CDCA1-LPs and the cross-priming potential of LPs bearing CTL-epitopes in both human in vitro and HLA-class I transgenic mice in vivo. Then we analyzed the Th cell response to CDCA1 in head-and-neck cancer (HNC) patients before and after vaccination with a CDCA1-derived CTL-epitope peptide using IFN-γ enzyme-linked immunospot assays. We identified two CDCA1-LPs, CDCA1(39­64)-LP and CDCA1(55­78)-LP, which encompass naturally processed epitopes recognized by Th cells and CTLs. CDCA1-specific CTLs were induced through cross-presentation of CDCA1-LPs in vitro and in vivo. In addition, CDCA1-specific Th cells enhanced induction of CDCA1-specific CTLs. Furthermore, significant frequencies of CDCA1-specific Th cell responses were detected after short-term in vitro stimulation of peripheral blood mononuclear cells (PBMCs) with CDCA1-LPs in HNC patients (CDCA1(39­64)-LP, 74%; CDCA1(55­78)-LP, 68%), but not in healthy donors. These are the first results demonstrating the presence of CDCA1-specific Th cell responses in HNC patients and underline the possible utility of CDCA1-LPs for propagation of both CDCA1-specific Th cells and CTLs.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Cycle Proteins/immunology , Head and Neck Neoplasms/immunology , Neoplasm Recurrence, Local/immunology , Peptide Fragments/immunology , Platelet Membrane Glycoprotein IIb/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Case-Control Studies , Cell Cycle Proteins/metabolism , Cells, Cultured , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Fibroblasts/immunology , Fibroblasts/metabolism , Fibroblasts/pathology , Flow Cytometry , HLA Antigens/immunology , HLA Antigens/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Mice , Neoplasm Metastasis , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Peptide Fragments/metabolism , Platelet Membrane Glycoprotein IIb/metabolism , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Th1 Cells/pathology
11.
J Leukoc Biol ; 114(1): 53-67, 2023 07 01.
Article in English | MEDLINE | ID: mdl-36976024

ABSTRACT

Despite effective antiretroviral therapy, HIV-1 persists in cells, including macrophages, which is an obstacle to cure. However, the precise role of macrophages in HIV-1 infection remains unclear because they reside in tissues that are not easily accessible. Monocyte-derived macrophages are widely used as a model in which peripheral blood monocytes are cultured and differentiated into macrophages. However, another model is needed because recent studies revealed that most macrophages in adult tissues originate from the yolk sac and fetal liver precursors rather than monocytes, and the embryonic macrophages possess a self-renewal (proliferating) capacity that monocyte-derived macrophages lack. Here, we show that human induced pluripotent stem cell-derived immortalized macrophage-like cells are a useful self-renewing macrophage model. They proliferate in a cytokine-dependent manner, retain macrophage functions, support HIV-1 replication, and exhibit infected monocyte-derived macrophage-like phenotypes, such as enhanced tunneling nanotube formation and cell motility, as well as resistance to a viral cytopathic effect. However, several differences are also observed between monocyte-derived macrophages and induced pluripotent stem cell-derived immortalized macrophage-like cells, most of which can be explained by the proliferation of induced pluripotent stem cell-derived immortalized macrophage-like cells. For instance, proviruses with large internal deletions, which increased over time in individuals receiving antiretroviral therapy, are enriched more rapidly in induced pluripotent stem cell-derived immortalized macrophage-like cells. Interestingly, inhibition of viral transcription by HIV-1-suppressing agents is more obvious in induced pluripotent stem cell-derived immortalized macrophage-like cells. Collectively, our present study proposes that the model of induced pluripotent stem cell-derived immortalized macrophage-like cells is suitable for mimicking the interplay between HIV-1 and self-renewing tissue macrophages, the newly recognized major population in most tissues that cannot be fully modeled by monocyte-derived macrophages alone.


Subject(s)
HIV Infections , HIV-1 , Induced Pluripotent Stem Cells , Adult , Humans , HIV-1/physiology , Macrophages , Monocytes , Cells, Cultured , Virus Replication
12.
ALTEX ; 40(2): 204-216, 2023.
Article in English | MEDLINE | ID: mdl-35229878

ABSTRACT

Although several in vitro assays that predict the sensitizing potential of chemicals have been developed, none can distinguish between chemical respiratory and skin sensitizers. Recently, we established a new three-dimensional dendritic cell (DC) coculture system consisting of a human airway epithelial cell line, immature DCs derived from human peripheral monocytes, and a human lung fibroblast cell line. In this coculture system, compared to skin sensitizers, respiratory sensitizers showed enhanced mRNA expression in DCs of the key costimulatory molecule OX40 ligand (OX40L), which is important for T helper 2 (Th2) cell differentiation. Herein, we established a new two-step DC/T cell coculture system by adding peripheral allogeneic naïve CD4+ T cells to the DCs stimulated in the DC coculture system. In this DC/T cell coculture system, model respiratory sensitizers, but not skin sensitizers, enhanced mRNA expression of the predominant Th2 marker interleukin-4 (IL-4). To improve the versatility, in place of peripheral monocytes, monocyte-derived proliferating cells called CD14-ML were used in the DC coculture system. As in peripheral monocytes, enhanced mRNA expression of OX40L was induced in CD14-ML by respiratory sensitizers compared to skin sensitizers. When these cell lines were applied to the DC/T cell coculture system with peripheral allogeneic naïve CD4+ T cells, respiratory sensitizers but not skin sensitizers enhanced the mRNA expression of IL-4. Thus, this DC/T cell coculture system may be useful for discriminating between respiratory and skin sensitizers by differential mRNA upregulation of IL-4 in T cells.


Subject(s)
Coculture Techniques , Interleukin-4 , Th2 Cells , Humans , Cell Differentiation , Cells, Cultured , Dendritic Cells , Interleukin-4/metabolism , Interleukin-4/pharmacology , Monocytes , RNA, Messenger/metabolism , Th2 Cells/metabolism
13.
FASEB J ; 25(8): 2757-69, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21515746

ABSTRACT

Eukaryotic mRNA turnover is among most critical mechanisms that affect mRNA abundance and are regulated by mRNA-binding proteins and the cytoplasmic exosome. A functional protein, guanosine-triphosphate-binding protein 1 (GTPBP1), which associates with both the exosome and target mRNAs, was identified. The overexpression of GTPBP1 accelerated the target mRNA decay, whereas the reduction of the GTPBP1 expression with RNA interference stabilized the target mRNA. GTPBP1 has a putative guanosine-triphosphate (GTP)-binding domain, which is found in members of the G-protein family and Ski7p, a well-known core factor of the exosome-mediated mRNA turnover pathway in yeast. Analyses of protein interactions and mRNA decay demonstrated that GTPBP1 modulates mRNA degradation via GTP-binding-dependent target loading. Moreover, GTPBP1-knockout models displayed multiple mRNA decay defects, including elevated nocturnal levels of Aanat mRNA in pineal glands, and retarded degradation of TNF-α mRNA in lipopolysaccharide-treated splenocytes. The results of this study suggest that GTPBP1 is a regulator and adaptor of the exosome-mediated mRNA turnover pathway.


Subject(s)
Exosomes/metabolism , Monomeric GTP-Binding Proteins/metabolism , RNA, Messenger/metabolism , 3' Untranslated Regions , Adaptor Proteins, Signal Transducing/metabolism , Animals , Base Sequence , Binding Sites/genetics , CHO Cells , Circadian Rhythm/genetics , Cricetinae , Cricetulus , DNA Primers/genetics , Guanosine Triphosphate/metabolism , HEK293 Cells , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , Humans , Mice , Mice, Inbred CBA , Mice, Knockout , Models, Biological , Monomeric GTP-Binding Proteins/deficiency , Monomeric GTP-Binding Proteins/genetics , Pineal Gland/metabolism , Protein Binding , RNA Stability , Rats , Rats, Sprague-Dawley , Saccharomyces cerevisiae Proteins/metabolism
14.
J Immunol ; 185(9): 5259-67, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20921531

ABSTRACT

TRAIL is known to play a pivotal role in the inhibition of autoimmune disease. We previously demonstrated that administration of dendritic cells engineered to express TRAIL and myelin-oligodendrocyte glycoprotein reduced the severity of experimental autoimmune encephalomyelitis and suggested that CD4(+)CD25(+) regulatory T cells (Tregs) were involved in mediating this preventive effect. In the current study, we investigated the effect of TRAIL on Tregs, as well as conventional T cells, using TRAIL-deficient mice. Upon induction of experimental autoimmune encephalomyelitis, TRAIL-deficient mice showed more severe clinical symptoms, a greater frequency of IFN-γ-producing CD4(+) T (Th1) cells, and a lower frequency of CD4(+)Foxp3(+) Tregs than did wild-type mice. In vitro, conventional T cells stimulated by bone marrow-derived dendritic cells (BM-DCs) from TRAIL-deficient mice showed a greater magnitude of proliferation than did those stimulated by BM-DCs from wild-type mice. In contrast, TRAIL expressed on the stimulator BM-DCs enhanced the proliferative response of CD4(+)CD25(+) Tregs in the culture. The functional TRAILR, mouse death receptor 5 (mDR5), was expressed in conventional T cells and Tregs upon stimulation. In contrast, the decoy receptor, mDc-TRAILR1, was slightly expressed only on CD4(+)CD25(+) Tregs. Therefore, the distinct effects of TRAIL may be due to differences in the mDc-TRAILR1 expression or the signaling pathways downstream of mouse death receptor 5 between the two T cell subsets. Our data suggest that TRAIL suppresses autoimmunity by two mechanisms: the inhibition of Th1 cells and the promotion of Tregs.


Subject(s)
Autoimmunity/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , TNF-Related Apoptosis-Inducing Ligand/immunology , Th1 Cells/immunology , Animals , Cell Proliferation , Cell Separation , Encephalomyelitis, Autoimmune, Experimental/immunology , Flow Cytometry , Immunohistochemistry , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/cytology , Th1 Cells/cytology , Tumor Necrosis Factor Decoy Receptors/immunology
15.
Cancer Sci ; 102(1): 71-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21087352

ABSTRACT

Insulin-like growth factor-II mRNA binding protein 3 (IMP-3) is an oncofetal protein expressed in various malignancies including lung cancer. This study aimed to identify immunogenic peptides derived from IMP-3 that can induce tumor-reactive and human leukocyte antigen (HLA)-A2 (A*02:01)-restricted cytotoxic T lymphocytes (CTL) for lung cancer immunotherapy. Forty human IMP-3-derived peptides predicted to bind to HLA-A2 were analyzed to determine their capacity to induce HLA-A2-restricted T cells in HLA-A2.1 (HHD) transgenic mice (Tgm). We found that three IMP-3 peptides primed HLA-A2-restricted CTL in the HLA-A2.1 Tgm. Among them, human CTL lines reactive to IMP-3 (515) NLSSAEVVV(523) were reproducibly established from HLA-A2-positive healthy donors and lung cancer patients. On the other hand, IMP-3 (199) RLLVPTQFV(207) reproducibly induced IMP-3-specific and HLA-A2-restricted CTL from healthy donors, but did not sensitize CTL in the HLA-A2.1 Tgm. Importantly, these two IMP-3 peptide-specific CTL generated from healthy donors and cancer patients effectively killed the cancer cells naturally expressing both IMP-3 and HLA-A2. Cytotoxicity was significantly inhibited by anti-HLA class I and anti-HLA-A2 monoclonal antibodies, but not by the anti-HLA-class II monoclonal antibody. In addition, natural processing of these two epitopes derived from the IMP-3 protein was confirmed by specific killing of HLA-A2-positive IMP-3-transfectants but not the parental IMP-negative cell line by peptide-induced CTL. This suggests that these two IMP-3-derived peptides represent highly immunogenic CTL epitopes that may be attractive targets for lung cancer immunotherapy.


Subject(s)
HLA-A2 Antigen/immunology , Neoplasms/immunology , Peptide Fragments/immunology , RNA-Binding Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Line, Tumor , Epitopes, T-Lymphocyte , Humans , Lung Neoplasms/immunology , Mice , RNA-Binding Proteins/genetics
16.
Cancer Sci ; 102(4): 697-705, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21231984

ABSTRACT

The present study attempted to identify a useful tumor-associated antigen (TAA) for lung cancer immunotherapy and potential immunogenic peptides derived from the TAA. We focused on cell division cycle 45-like (CDC45L), which has a critical role in the initiation and elongation steps of DNA replication, as a novel candidate TAA for immunotherapy based on a genome-wide cDNA microarray analysis of lung cancer. The CDC45L was overexpressed in the majority of lung cancer tissues, but not in the adjacent non-cancerous tissues or in many normal adult tissues. We examined the in vitro and in vivo anti-tumor effects of cytotoxic T-lymphocytes (CTL) specific to CDC45L-derived peptides induced from HLA-A24 (A*24:02)-positive donors. We identified three CDC45L-derived peptides that could reproducibly induce CDC45L-specific and HLA-A24-restricted CTL from both healthy donors and lung cancer patients. The CTL could effectively lyse lung cancer cells that endogenously expressed both CDC45L and HLA-A24. In addition, we found that CDC45L (556) KFLDALISL(564) was eminent in that it induced not only HLA-A24 but also HLA-A2 (A*02:01)-restricted antigen specific CTL. Furthermore, the adoptive transfer of the CDC45L-specific CTL inhibited the growth of human cancer cells engrafted into immunocompromised mice. These results suggest that these three CDC45L-derived peptides are highly immunogenic epitopes and CDC45L is a novel TAA that might be a useful target for lung cancer immunotherapy.


Subject(s)
Cell Cycle Proteins/metabolism , Epitopes/immunology , HLA-A Antigens/immunology , Neoplasms/immunology , Neoplasms/therapy , Peptide Fragments/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Northern , Blotting, Western , Cell Cycle Proteins/genetics , Cell Cycle Proteins/immunology , Gene Expression Profiling , HLA-A Antigens/metabolism , HLA-A24 Antigen , Humans , Immunoenzyme Techniques , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms/metabolism , Oligonucleotide Array Sequence Analysis , Peptide Fragments/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
17.
J Immunol ; 183(1): 201-8, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19542431

ABSTRACT

CD1d-restricted invariant NKT (iNKT) cells play crucial roles in various types of immune responses, including autoimmune diseases, infectious diseases and tumor surveillance. The mechanisms underlying their adjuvant functions are well understood. Nevertheless, although IL-4 and IL-10 production characterize iNKT cells able to prevent or ameliorate some autoimmune diseases and inflammatory conditions, the precise mechanisms by which iNKT cells exert immune regulatory function remain elusive. This study demonstrates that the activation of human iNKT cells by their specific ligand alpha-galactosylceramide enhances IL-12p70 while inhibiting the IL-23 production by monocyte-derived dendritic cells, and in turn down-regulating the IL-17 production by memory CD4(+) Th cells. The ability of the iNKT cells to regulate the differential production of IL-12p70/IL-23 is mainly mediated by a remarkable hallmark of their function to produce both Th1 and Th2 cytokines. In particular, the down-regulation of IL-23 is markedly associated with a production of IL-4 and IL-10 from iNKT cells. Moreover, Th2 cytokines, such as IL-4 and IL-13 play a crucial role in defining the biased production of IL-12p70/IL-23 by enhancement of IL-12p70 in synergy with IFN-gamma, whereas inhibition of the IFN-gamma-promoted IL-23 production. Collectively, the results suggest that iNKT cells modify the IL-12p70/IL-23 balance to enhance the IL-12p70-induced cell-mediated immunity and suppress the IL-23-dependent inflammatory pathologies. These results may account for the long-appreciated contrasting beneficial and adverse consequence of ligand activation of iNKT cells.


Subject(s)
Cytokines/physiology , Dendritic Cells/immunology , Interleukin-12/metabolism , Interleukin-23/metabolism , Lymphocyte Activation/immunology , Natural Killer T-Cells/immunology , Protein Subunits/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Cells, Cultured , Dendritic Cells/metabolism , Galactosylceramides/metabolism , Humans , Interleukin-12/biosynthesis , Interleukin-23/antagonists & inhibitors , Interleukin-23/biosynthesis , Ligands , Natural Killer T-Cells/metabolism , Protein Subunits/biosynthesis , Receptors, Antigen, T-Cell/metabolism
18.
Int J Cancer ; 126(9): 2153-63, 2010 May 01.
Article in English | MEDLINE | ID: mdl-19688828

ABSTRACT

The present study attempted to identify a target antigen for immunotherapy for cholangiocarcinoma. Forkhead box M1 (FOXM1) was selected as a candidate antigen based on the data of previous cDNA microarray analysis of clinical samples of cholangiocarcinoma. The level of FOXM1 mRNA was more than 4 times higher in cancer cells in comparison to adjacent normal epithelial cells, in all of 24 samples of cholangiocarcinoma tissues. An immunohistochemical analysis also detected FOXM1 protein in the cancer cells but not in the normal cells. Twenty-three human FOXM1-derived peptides predicted to bind to HLA-A2 were analyzed to determine their ability to induce HLA-A2-restricted T cells in HLA-A2 transgenic mice. FOXM1(362-370) (YLVPIQFPV), FOXM1(373-382) (SLVLQPSVKV), and FOXM1(640-649) (GLMDLSTTPL) peptides primed HLA-A2-restricted cytotoxic T lymphocytes (CTLs) in the HLA-A2 transgenic mice. Human CTL lines reactive to these 3 peptides could also be established from HLA-A2-positive healthy donors and cancer patients. Natural processing of the 3 epitopes from FOXM1 protein was confirmed by specific killing of HLA-A2-positive FOXM1-transfectants by peptide-induced CTLs. FOXM1 is expressed in various types of cancers and it is also functionally involved in oncogenic transformation and the survival of cancer cells. Therefore, FOXM1 may be a suitable target for immunotherapy against various cancers including cholangiocarcinoma.


Subject(s)
Forkhead Transcription Factors/immunology , Immunotherapy , Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Animals , Bile Duct Neoplasms/therapy , Bile Ducts, Intrahepatic , Cell Line, Tumor , Cholangiocarcinoma/therapy , Epitopes, T-Lymphocyte , Forkhead Box Protein M1 , Forkhead Transcription Factors/analysis , Forkhead Transcription Factors/genetics , HLA-A2 Antigen/immunology , Humans , Mice , Mice, Transgenic , Neoplasms/immunology , Oligonucleotide Array Sequence Analysis , Peptide Fragments/immunology , RNA, Messenger/analysis
19.
Int J Cancer ; 127(6): 1393-403, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20063317

ABSTRACT

To establish efficient anticancer immunotherary, it is important to identify tumor-associated antigens (TAAs) directing the immune system to attack cancer. A genome-wide cDNA microarray analysis identified that secreted protein acidic and rich in cysteine (SPARC) gene is overexpressed in the gastric, pancreatic and colorectal cancer tissues but not in their noncancerous counterparts. This study attempted to identify HLA-A24 (A*2402)-restricted and SPARC-derived CTL epitopes. We previously identified H-2K(d)-restricted and SPARC-derived CTL epitope peptides in BALB/c mice, of which H-2K(d)-binding peptide motif is comparable with that of HLA-A24 binding peptides. By using these peptides, we tried to induce HLA-A24 (A*2402)-restricted and SPARC-reactive human CTLs and demonstrated an antitumor immune response. The SPARC-A24-1(143-151) (DYIGPCKYI) and SPARC-A24-4(225-234) (MYIFPVHWQF) peptides-reactive CTLs were successfully induced from peripheral blood mononuclear cells by in vitro stimulation with these two peptides in HLA-A24 (A*2402) positive healthy donors and cancer patients, and these CTLs exhibited cytotoxicity specific to cancer cells expressing both SPARC and HLA-A24 (A*2402). Furthermore, the adoptive transfer of the SPARC-specific CTLs could inhibit the tumor growth in nonobese diabetic/severe combined immunodeficient mice bearing human cancer cells expressing both HLA-A24 (A*2402) and SPARC. These findings suggest that SPARC is a potentially useful target candidate for cancer immunotherapy.


Subject(s)
Immunotherapy, Adoptive , Neoplasms/therapy , Osteonectin/immunology , Amino Acid Sequence , Animals , Base Sequence , DNA Primers , Female , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Neoplasms/immunology , Oligonucleotide Array Sequence Analysis , Osteonectin/chemistry , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes, Cytotoxic/immunology
20.
Stem Cells ; 27(5): 1021-31, 2009 May.
Article in English | MEDLINE | ID: mdl-19415766

ABSTRACT

Methods have been established to generate dendritic cells (DCs) from mouse and human embryonic stem (ES) cells. We designated them as ES-DCs and mouse models have demonstrated the induction of anti-cancer immunity and prevention of autoimmune disease by in vivo administration of genetically engineered ES-DCs. For the future clinical application of ES-DCs, the histoincompatibility between patients to be treated and available human ES cells and the ethical concerns associated with human ES cells may be serious obstacles. However, recently developed induced pluripotent stem (iPS) cell technology is expected to resolve these issues. This report describes the generation and characterization of DCs derived from mouse iPS cells. The iPS cell-derived DCs (iPS-DCs) possessed the characteristics of DCs including the capacity of T-cell-stimulation, antigen-processing and presentation and cytokine production. DNA microarray analyses revealed the upregulation of genes related to antigen-presenting functions during differentiation into iPS-DCs and similarity in gene expression profile in iPS-DCs and bone marrow cell-derived DCs. Genetically modified iPS-DCs expressing antigenic protein primed T-cells specific to the antigen in vivo and elicited efficient antigen-specific anti-tumor immunity. In addition, macrophages were generated from iPS cells (iPS-MP). iPS-MP were comparable with bone marrow cell-derived macrophages in the cell surface phenotype, functions, and gene expression profiles.


Subject(s)
Cell Differentiation , Dendritic Cells/cytology , Macrophages/cytology , Pluripotent Stem Cells/cytology , Animals , Antigen Presentation/immunology , Cell Line , Cell Shape , Cross-Priming/immunology , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Epitopes/immunology , Gene Expression Profiling , Gene Expression Regulation , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Phenotype , Pluripotent Stem Cells/metabolism , Surface Properties , T-Lymphocytes/cytology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL