Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
bioRxiv ; 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37961107

ABSTRACT

Effector T helper (Th) cell differentiation is fundamental to functional adaptive immunity. Different subsets of dendritic cells (DCs) preferentially induce different types of Th cells, but the fate instruction mechanism for Th type 2 (Th2) differentiation remains enigmatic, as the critical DC-derived cue has not been clearly identified. Here, we show that CD301b+ DCs, a major Th2-inducing DC subset, drive Th2 differentiation through cognate interaction by 'kick-starting' IL-2 receptor signaling in CD4T cells. Mechanistically, CD40 engagement induces IL-2 production selectively from CD301b+ DCs to maximize CD25 expression in CD4 T cells, which is required specifically for the Th2 fate decision. On the other hand, CD25 in CD301b+ DCs facilitates directed action of IL-2 toward cognate CD4T cells. Furthermore, CD301b+ DC-derived IL-2 skews CD4T cells away from the T follicular helper fate. These results highlight the critical role of DC-intrinsic CD40-IL-2 axis in bifurcation of Th cell fate.

2.
Curr Opin Immunol ; 83: 102352, 2023 08.
Article in English | MEDLINE | ID: mdl-37276821

ABSTRACT

Conventional dendritic cells (cDCs) are potent antigen-presenting cells that consist of developmentally, phenotypically, and functionally distinct subsets. Following immunization, each subset of cDCs acquires the antigen and presents it to CD4T (CD4+ T (cells)) cells with distinct spatiotemporal kinetics in the secondary lymphoid organs, often causing multiple waves of antigen presentation to CD4T cells. Here, we review the current understanding of the kinetics of antigen presentation by each cDC subset and its functional consequences in priming naive CD4T cells, and discuss its implications in the differentiation of CD4T cells.


Subject(s)
CD4-Positive T-Lymphocytes , Dendritic Cells , Mice , Animals , Antigen Presentation , Antigens
3.
STAR Protoc ; 3(4): 101845, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36595900

ABSTRACT

Physical contact between T cells and antigen-presenting cells (APCs) is essential for priming antigen-specific T cells, but quantitating the antigen-dependent T cell-APC contact can be laborious. Here, we present a simple flow-cytometry-based protocol for quantitating T cell-APC contacts in the antigen-draining lymph node in mice immunized with ovalbumin (OVA). This protocol quantifies the contact between adoptively transferred OVA-specific TCR transgenic CD4T (OT-II) cells and dendritic cell (DC) subsets. This approach can be applied to other types of intercellular interactions between T cells and APCs. For complete details on the use and execution of this protocol, please refer to Tatsumi et al. (2021).1.


Subject(s)
Antigens , T-Lymphocytes , Mice , Animals , Flow Cytometry , Antigen-Presenting Cells , Lymph Nodes/pathology
4.
Sci Immunol ; 6(66): eabg0336, 2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34890253

ABSTRACT

During the initiation of adaptive immune responses, millions of lymphocytes must be scanned to find the few cognate clones. The activation mechanisms of CD4 T cells have been extensively studied, but the cellular mechanisms that drive selection of cognate clones are not completely understood. Here, we show that recently homed naïve polyclonal CD4 T cells are temporarily retained before leaving the lymph node. This stop-and-go traffic of CD4 T cells provides an adequate time window for efficient scanning and timely priming of antigen-specific cognate clones. CD301b+ DCs, a major subset of migratory cDC2 cells, localize to the areas around high endothelial venules, where they retain incoming polyclonal CD4 T cells through MHCII-dependent but antigen-independent mechanisms, while concurrently providing cognate stimuli for priming. These results indicate that CD301b+ DCs function as an immunological "display window" for CD4 T cells to efficiently scan their antigen specificity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Lectins, C-Type/immunology , Lymph Nodes/immunology , Animals , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
5.
J Immunother ; 39(3): 127-39, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26938946

ABSTRACT

Many studies demonstrated crucial roles of avidity of T-cell receptor (TCR) in T-cell fate. However, majority of these findings resulted from analysis of non-self-antigen-specific CD8 T cells, and little is known about roles of TCR avidity in the fate of self-antigen-specific CD8 T cells. Wilms tumor gene 1 (WT1) protein is a self-antigen most suitable for addressing this issue because WT1 protein is a highly immunogenic, typical self-antigen. Here, we isolated 2 distinct and functional TCRs, TCR1 and TCR2, from murine WT1 peptide (RMFPNAPYL)-specific cytotoxic T lymphocytes (WT1-CTLs) and generated TCR1-retrogenic (Rg) and TCR2-Rg mice under T and B-cell-deficient and -reconstituted conditions. TCR1-transduced CD8 T (TCR1-T) cells had approximately 2-fold higher avidity to WT1 peptide than TCR2-transduced CD8 T (TCR2-T) cells. Cytokine production profiles and cell surface phenotypes showed that TCR1-T cells were more differentiated than TCR2-T cells under both conditions. Therefore, TCR1-T cells with TCR avidity higher than that of TCR2-T cells are more differentiated compared with TCR2-T cells. Furthermore, TCR1-T cells that developed under T and B-cell-reconstituted conditions displayed cytotoxicity against endogenously WT1-expressing tumor cells, whereas TCR2 T cells that developed under the same conditions did not. Thus, it was demonstrated, for the first time, that TCR avidity played an essential role in differentiation of self-antigen-reactive T cells, through the success of establishment of two distinct WT1-CTLs with a difference in only TCR avidity under the identical genetic background. Present results should provide us with an insight for elucidation of the differentiation mechanisms of self-antigen-reactive T cells, including tumor antigen-reactive T cells.


Subject(s)
Autoantigens/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Autoantigens/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line , Cloning, Molecular , Cytokines/biosynthesis , Cytotoxicity, Immunologic/immunology , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Gene Expression , Immunophenotyping , Lymphocyte Activation , Mice , Mice, Transgenic , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/metabolism , Peptide Fragments/immunology , Peptide Fragments/metabolism , Phenotype , Protein Binding/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Transduction, Genetic , WT1 Proteins/chemistry , WT1 Proteins/genetics , WT1 Proteins/immunology , WT1 Proteins/metabolism
6.
PLoS One ; 11(3): e0152326, 2016.
Article in English | MEDLINE | ID: mdl-27011118

ABSTRACT

To improve cancer immunotherapy, it is important to understand how tumor cells counteract immune-surveillance. In this study, we sought to identify cell-surface molecules associated with resistance of leukemia cells to cytotoxic T cell (CTL)-mediated cytolysis. To this end, we first established thousands of monoclonal antibodies (mAbs) that react with MLL/AF9 mouse leukemia cells. Only two of these mAbs, designated R54 and B2, bound preferentially to leukemia cells resistant to cytolysis by a tumor cell antigen-specific CTLs. The antigens recognized by these mAbs were identified by expression cloning as the same protein, CD43, although their binding patterns to subsets of hematopoietic cells differed significantly from each other and from a pre-existing pan-CD43 mAb, S11. The epitopes of R54 and B2, but not S11, were sialidase-sensitive and expressed at various levels on leukemia cells, suggesting that binding of R54 or B2 is associated with the glycosylation status of CD43. R54high leukemia cells, which are likely to express sialic acid-rich CD43, were highly resistant to CTL-mediated cytolysis. In addition, loss of CD43 in leukemia cells or neuraminidase treatment of leukemia cells sensitized leukemia cells to CTL-mediated cell lysis. These results suggest that sialic acid-rich CD43, which harbors multiple sialic acid residues that impart a net negative surface charge, protects leukemia cells from CTL-mediated cell lysis. Furthermore, R54high or B2high leukemia cells preferentially survived in vivo in the presence of adaptive immunity. Taken together, these results suggest that the glycosylation status of CD43 on leukemia is associated with sensitivity to CTL-mediated cytolysis in vitro and in vivo. Thus, regulation of CD43 glycosylation is a potential strategy for enhancing CTL-mediated immunotherapy.


Subject(s)
Drug Resistance, Neoplasm , Leukemia/immunology , Leukemia/metabolism , Leukosialin/chemistry , Adaptive Immunity , Animals , Bone Marrow Cells/cytology , Cell Line, Tumor , Cell Separation , Cytotoxicity, Immunologic , Epitopes/chemistry , Female , Flow Cytometry , Gene Expression Regulation, Leukemic , Glycosylation , Humans , Immunotherapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , N-Acetylneuraminic Acid/chemistry , Rats , Rats, Inbred Lew , Stem Cells/cytology , T-Lymphocytes, Cytotoxic/cytology
7.
Oncol Rep ; 35(4): 1959-66, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26820500

ABSTRACT

Eukaryotic elongation factor 2 (eEF2) is an essential factor for protein synthesis. Previous studies have shown that the eEF2 gene was overexpressed and plays an oncogenic role in various types of cancers and that eEF2 gene product elicited both humoral immune responses to produce eEF2-specific IgG autoantibody in cancer-bearing individuals and cellular immune responses to induce eEF2 peptide-specific cytotoxic T lymphocytes (CTLs) in vitro. The purpose of the present study was to induce eEF2-specific, antitumor CTL responses in vivo by vaccination with MHC class I-binding eEF2-derived peptide. First, two mouse MHC class I-restricted eEF2­derived, 9-mer peptides, EF17 (17-25 aa, ANIRNMSVI) and EF180 (180-188 aa, RIVENVNVI) were identified as eEF2-specific CTL peptides, and mice were vaccinated intradermally eight times with either EF17 or EF180 peptide emulsified with Montanide ISA51 adjuvant. Cytotoxicity assay showed that eEF2-specific CTLs were induced in both EF17­and EF180­vaccinated mice, and histological study showed no detectable damage in the organs of these mice. Next, to examine in vivo antitumor effects of eEF2 peptide vaccination in a therapeutic model, mice were vaccinated four times with one each of the two eEF2 peptides at weekly intervals after implantation of eEF2-expressing leukemia cells. The vaccination with eEF2 peptides induced eEF2-specific CTLs and suppressed tumor growth, and disease-free survival was significantly longer in EF180-vaccinated mice compared to control mice. The survival was associated with the robustness of eEF2-specific CTL induction. These results indicate that vaccination with MHC class I-binding eEF2 peptide induced eEF2-targeting, antitumor CTL responses in vivo without damage to normal organs, which provided us a rationale for eEF2 peptide-based cancer immunotherapy.


Subject(s)
Cancer Vaccines/immunology , Immunologic Factors/administration & dosage , Leukemia/prevention & control , Peptide Elongation Factor 2/immunology , Peptide Fragments/administration & dosage , T-Lymphocytes, Cytotoxic/immunology , Animals , Cancer Vaccines/administration & dosage , Cell Line, Tumor , Cytotoxicity, Immunologic , Leukemia/immunology , Male , Mice , Xenograft Model Antitumor Assays
8.
Future Sci OA ; 2(4): FSO96, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28116121

ABSTRACT

AIM: In cancer immunotherapy, biomarkers are important for identification of responsive patients. This study was aimed to find biomarkers that predict clinical outcome of WT1 peptide vaccination. MATERIALS & METHODS: Candidate genes that were expressed differentially between long- and short-term survivors were identified by cDNA microarray analysis of peripheral blood mononuclear cells that were extracted from 30 glioblastoma patients (discovery set) prior to vaccination and validated by quantitative RT-PCR using discovery set and different 23 patients (validation set). RESULTS: SDC-4 mRNA expression levels distinguished between the long- and short-term survivors: 1-year survival rates were 64.0 and 18.5% in SDC4-low and -high patients, respectively. CONCLUSION: SDC-4 is a novel predictive biomarker for the efficacy of WT1 peptide vaccine.

9.
Mol Carcinog ; 55(12): 2001-2009, 2016 12.
Article in English | MEDLINE | ID: mdl-26713860

ABSTRACT

In our previous study, we showed that miR-125a directly targeted a WT1 oncogene, which was overexpressed in leukemia and various kinds of solid tumors including lung, breast, gastric, and colon cancers, and brain tumors and was deeply involved in leukemogenesis and tumorigenesis and that miR-125a knockout mice overexpressed WT1 and developed myeloproliferative disease. It had been also reported that miR-125a is downregulated in leukemia and various types of solid tumors such as lung cancers, suggesting its tumor suppressor function. Therefore, it is important to elucidate what is target(s) of miR-125a for understandings of such functions although few target genes for it are known. In the present study, Zbtb7a oncogene was identified as a potential target for miR-125a by gene expression profiling in miR-125a knockout mice combined with bioinformatics target prediction. EGFP-3'UTR reporter assay showed that miR-125a suppressed Zbtb7a expression through its direct binding to the Zbtb7a-3'UTR. Zbtb7a knockdown by siRNA suppressed cell proliferation and induced G1 cell cycle arrest and apoptosis in lung cancer cells. Furthermore, miR-125a expression showed a negative correlation with Zbtb7a expression in non-small cell lung cancer tissues. The present study showed for the first time that Zbtb7a was a direct target for miR-125a and was involved in cell cycle progression and apoptosis of lung cancer cells. These results also demonstrated that deregulation of miR-125a-Zbtb7a signaling was associated with the development and progression of lung cancer. © 2015 Wiley Periodicals, Inc.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , MicroRNAs/genetics , Transcription Factors/genetics , Animals , Apoptosis , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Humans , Lung/metabolism , Lung/pathology , Lung Neoplasms/pathology , Mice, Knockout , Proto-Oncogene Mas
10.
PLoS One ; 10(12): e0144594, 2015.
Article in English | MEDLINE | ID: mdl-26658107

ABSTRACT

Leukemia differs substantially with respect to stromal milieu from tumors that progress locally as solid masses, and the physiological importance of immunosurveillance in leukemia remains unclear. However, currently available mouse leukemia models have critical limitations in the context of analyzing immunological regulation of leukemia development. In this study, we transferred mouse MLL/AF9 leukemia-initiating cells into immunocompetent recipient mice without any pre-conditioning such as irradiation, and then analyzed the spontaneous T cell response to an immunogenic antigen expressed in leukemia cells. When the minimum numbers of leukemia-initiating cells for engraftment were transferred, leukemia cells were eradicated by the adaptive immune response in most, if not all, wild-type mice, but not in Rag2-/- recipient mice, which lack adaptive immunity. By contrast, mice transplanted with larger numbers of leukemia cells always developed leukemia. In mice with advanced leukemia, antigen-specific CTLs were also expanded, but were unresponsive to antigen stimulation and expressed high levels of PD-1 and LAG-3. These results provide the first clear demonstration that the spontaneous CTL response to a tumor-cell antigen has the potential to eradicate leukemia, whereas antigen-specific CTLs are exhausted in animals with advanced leukemia. This immunocompetent mouse leukemia model provides a useful platform for developing effective immunotherapies against leukemia.


Subject(s)
Immunocompetence , Leukemia/immunology , Myeloid-Lymphoid Leukemia Protein/metabolism , T-Lymphocytes, Cytotoxic/immunology , Adaptive Immunity , Animals , Antigens, Neoplasm/immunology , Cell Line, Tumor , Disease Models, Animal , Disease Progression , Female , Leukemia/pathology , Mice, Inbred C57BL , Mice, Transgenic
11.
PLoS One ; 10(6): e0130578, 2015.
Article in English | MEDLINE | ID: mdl-26090994

ABSTRACT

The Wilms' tumor gene WT1 consists of 10 exons and encodes a zinc finger transcription factor. There are four major WT1 isoforms resulting from alternative splicing at two sites, exon 5 (17AA) and exon 9 (KTS). All major WT1 isoforms are overexpressed in leukemia and solid tumors and play oncogenic roles such as inhibition of apoptosis, and promotion of cell proliferation, migration and invasion. In the present study, a novel alternatively spliced WT1 isoform that had an extended exon 4 (designated as exon 4a) with an additional 153 bp (designated as 4a sequence) at the 3' end was identified and designated as an Ex4a(+)WT1 isoform. The insertion of exon 4a resulted in the introduction of premature translational stop codons in the reading frame in exon 4a and production of C-terminal truncated WT1 proteins lacking zinc finger DNA-binding domain. Overexpression of the truncated Ex4a(+)WT1 isoform inhibited the major WT1-mediated transcriptional activation of anti-apoptotic Bcl-xL gene promoter and induced mitochondrial damage and apoptosis. Conversely, suppression of the Ex4a(+)WT1 isoform by Ex4a-specific siRNA attenuated apoptosis. These results indicated that the Ex4a(+)WT1 isoform exerted dominant negative effects on anti-apoptotic function of major WT1 isoforms. Ex4a(+)WT1 isoform was endogenously expressed as a minor isoform in myeloid leukemia and solid tumor cells and increased regardless of decrease in major WT1 isoforms during apoptosis, suggesting the dominant negative effects on anti-apoptotic function of major WT1 isoforms. These results indicated that Ex4a(+)WT1 isoform had an important physiological function that regulated oncogenic function of major WT1 isoforms.


Subject(s)
WT1 Proteins/chemistry , WT1 Proteins/metabolism , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Base Sequence , Cloning, Molecular , Doxorubicin/toxicity , Exons , HL-60 Cells , Haplorhini , Humans , K562 Cells , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , RNA Interference , RNA, Small Interfering/metabolism , Sequence Analysis, DNA , WT1 Proteins/antagonists & inhibitors , WT1 Proteins/genetics
12.
Anticancer Res ; 35(3): 1251-61, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25750272

ABSTRACT

BACKGROUND/AIM: Wilms' tumor gene 1 (WT1) product is a pan-tumor-associated antigen. We previously identified WT1 protein-derived promiscuous helper peptide, WT1332. Therefore, isolation and characterization of the WT1332-specific T-cell receptors (TCRs) are useful to develop broadly applicable TCR gene-based adoptive immunotherapy. MATERIALS AND METHODS: A novel HLA-DRB1*04:05-restricted WT1332-specific TCR gene was cloned and transduced into human CD4+ T-cells by using a lentiviral vector. RESULTS: The WT1332-specific TCR-transduced CD4+ T-cells showed strong proliferation and Th1-cytokine production in an HLA-DRB1*04:05-restricted, WT1332-specific manner. Furthermore, the WT1332-specific TCR-transduced CD4+ T-cells could lyse HLA-DRB1*04:05-positive, WT1-expressing leukemia cells in vitro. CONCLUSION: The novel TCR gene cloned here should be a promising tool to develop adoptive immunotherapy of WT1332-specific TCR-transduced CD4+ T-cells for the treatment of WT1-expressing cancer, such as leukemia.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Genes, T-Cell Receptor , HLA-DRB1 Chains/immunology , Immunotherapy, Adoptive , Leukemia/therapy , WT1 Proteins/immunology , Amino Acid Sequence , Cloning, Molecular , Cytokines/analysis , Cytotoxicity, Immunologic , Humans , Jurkat Cells , Molecular Sequence Data , Transduction, Genetic
13.
Mol Carcinog ; 54(12): 1758-71, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25418835

ABSTRACT

The Wilms' tumor gene WT1 is overexpressed in leukemia and various types of solid tumors and plays an oncogenic role in these malignancies. Alternative splicing at two sites yields four major isoforms, 17AA(+)KTS(+), 17AA(+)KTS(-), 17AA(-)KTS(+), and 17AA(-)KTS(-), and all the isoforms are expressed in the malignancies. However, among the four isoforms, function of WT1[17AA(-)KTS(+)] isoform still remains undetermined. In the present study, we showed that forced expression of WT1[17AA(-)KTS(+)] isoform significantly inhibited apoptosis by DNA-damaging agents such as Doxorubicin, Mitomycin, Camptothesisn, and Bleomycin in immortalized fibroblast MRC5SV and cervical cancer HeLa cells. Knockdown of Rad51, an essential factor for homologous recombination (HR)-mediated DNA repair canceled the resistance to Doxorubicin induced by WT1[17AA(-)KTS(+)] isoform. GFP recombination assay showed that WT1[17AA(-)KTS(+)] isoform alone promoted HR, but that three other WT1 isoforms did not. WT1[17AA(-)KTS(+)] isoform significantly upregulated the expression of HR genes, XRCC2, Rad51D, and Rad54. Knockdown of XRCC2, Rad51D, and Rad54 inhibited the HR activity and canceled resistance to Doxorubicin in MRC5SV cells with forced expression of WT1[17AA(-)KTS(+)] isoform. Furthermore, chromatin immunoprecipitation (ChIP) assay showed the binding of WT1[17AA(-)KTS(+)] isoform protein to promoters of XRCC2 and Rad51D. Immunohistochemical study showed that Rad54 and XRCC2 proteins were highly expressed in the majority of non-small-cell lung cancer (NSCLC) and gastric cancer, and that expression of these two proteins was significantly correlated with that of WT1 protein in NSCLCs. Our results presented here showed that WT1[17AA(-)KTS(+)] isoform had a function to promote HR-mediated DNA repair.


Subject(s)
DNA Damage/genetics , DNA Repair/genetics , Genes, Wilms Tumor/physiology , Homologous Recombination/genetics , WT1 Proteins/genetics , Alternative Splicing/genetics , Apoptosis/genetics , Carcinoma, Non-Small-Cell Lung/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , HeLa Cells , Humans , Lung Neoplasms/genetics , Nuclear Proteins/genetics , Promoter Regions, Genetic/genetics , Protein Isoforms/genetics , Stomach Neoplasms/genetics
14.
Int J Oncol ; 44(5): 1461-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24589652

ABSTRACT

Recent studies have shown that cancer immunotherapy could be a promising therapeutic approach for the treatment of cancer. In the present study, to identify novel tumor-associated antigens (TAAs), the proteins expressed in a panel of cancer cells were serologically screened by immunoblot analysis and the eukaryotic elongation factor 2 (eEF2) was identified as an antigen that was recognized by IgG autoantibody in sera from a group of patients with head and neck squamous cell carcinoma (HNSCC) or colon cancer. Enzyme-linked immunosorbent assay showed that serum eEF2 IgG Ab levels were significantly higher in colorectal and gastric cancer patients compared to healthy individuals. Immunohistochemistry experiments showed that the eEF2 protein was overexpressed in the majority of lung, esophageal, pancreatic, breast and prostate cancers, HNSCC, glioblastoma multiforme and non-Hodgkin's lymphoma (NHL). Knockdown of eEF2 by short hairpin RNA (shRNA) significantly inhibited the growth in four eEF2-expressing cell lines, PC14 lung cancer, PCI6 pancreatic cancer, HT1080 fibrosarcoma and A172 glioblastoma cells, but not in eEF2-undetectable MCF7 cells. Furthermore, eEF2-derived 9-mer peptides, EF786 (eEF2 786-794 aa) and EF292 (eEF2 292-300 aa), elicited cytotoxic T lymphocyte (CTL) responses in peripheral blood mononuclear cells (PBMCs) from an HLA-A*24:02- and an HLA-A*02:01-positive healthy donor, respectively, in an HLA-A-restricted manner. These results indicated that the eEF2 gene is overexpressed in the majority of several types of cancers and plays an oncogenic role in cancer cell growth. Moreover, the eEF2 gene product is immunogenic and a promising target molecule of cancer immunotherapy for several types of cancers.


Subject(s)
Antigens, Neoplasm/genetics , Elongation Factor 2 Kinase/genetics , Neoplasms/genetics , Neoplasms/immunology , Antigens, Neoplasm/metabolism , Cell Line, Tumor , Elongation Factor 2 Kinase/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Immunoglobulin G/immunology , MCF-7 Cells , Neoplasms/pathology , Sequence Analysis, DNA , T-Lymphocytes, Cytotoxic/immunology
15.
J Immunother ; 36(3): 159-70, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23502763

ABSTRACT

Wilms tumor gene 1 (WT1) is overexpressed in various malignant neoplasms, and has been demonstrated as an attractive target for cancer immunotherapy. We previously reported the identification of a WT1 protein-derived, 16-mer helper peptide WT1332 that could elicit Th1-type CD4+ T-cell response and bind to multiple HLA class II molecules. In this study, we examined the feasibility of adoptive therapy using CD4+ T cells that were transduced an HLA-DPB1*05:01-restricted, WT1332-specific T-cell receptor (TCR). HLA-DPB1*05:01-restricted, WT1332-specific TCR-transduced CD4+ T cells were successfully generated using lentiviral vector and exhibited strong proliferative response and Th1-type cytokine production in response to WT1332 peptide, WT1 protein, or WT1-expressing tumor cell lysate. Furthermore, the WT1332-specific TCR-transduced CD4+ T cells lysed HLA-DPB1*05:01-positive, WT1-expressing human leukemia cells through granzyme B/perforin pathway. Furthermore, stimulation of peripheral blood mononuclear cells with both HLA-A*24:02-restricted cytotoxic T lymphocytes-epitope peptide (modified 9-mer WT1235 peptide, WT1235m) and WT1332 helper peptide in the presence of WT1332-specific TCR-transduced CD4+ T cells strikingly enhanced the induction of WT1235m-specific cytotoxic T lymphocytes. Thus, these results demonstrated the feasibility of immunotherapy based on adoptive transfer of WT1332-specific TCR-transduced CD4+ T cells for the treatment of leukemia.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HLA-DP beta-Chains/immunology , Leukemia/immunology , T-Lymphocytes, Cytotoxic/immunology , WT1 Proteins/immunology , Animals , Cell Line , Cell Proliferation , HLA-A24 Antigen/immunology , Humans , Immunotherapy, Adoptive , Leukocytes, Mononuclear/immunology , Mice , Receptors, Antigen, T-Cell/immunology
16.
Vaccine ; 30(4): 722-9, 2012 Jan 17.
Article in English | MEDLINE | ID: mdl-22133512

ABSTRACT

To induce and activate tumor-associated antigen-specific cytotoxic T lymphocytes (CTLs) for cancer immunity, it is important not only to select potent CTL epitopes but also to combine them with appropriate immunopotentiating agents. Here we investigated whether tumor immunity induced by WT1 peptide vaccination could be enhanced by IFN-ß. For the experimental group, C57BL/6 mice were twice pre-treated with WT1 peptide vaccine, implanted with WT1-expressing C1498 cells, and treated four times with WT1 peptide vaccine at one-week intervals. During the vaccination period, IFN-ß was injected three times a week. Mice in control groups were treated with WT1 peptide alone, IFN-ß alone, or PBS alone. The mice in the experimental group rejected tumor cells and survived significantly longer than mice in the control groups. The overall survival on day 75 was 40% for the mice treated with WT1 peptide+IFN-ß, while it was 7, 7, and 0% for those treated with WT1 peptide alone, IFN-ß alone or PBS alone, respectively. Induction of WT1-specific CTLs and enhancement of NK activity were detected in splenocytes from mice in the experimental group. Furthermore, administration of IFN-ß enhanced expression of MHC class I molecules on the implanted tumor cells. In conclusion, our results showed that co-administration of WT1 peptide+IFN-ß enhanced tumor immunity mainly through the induction of WT1-specific CTLs, enhancement of NK activity, and promotion of MHC class I expression on the tumor cells. WT1 peptide vaccination combined with IFN-ß administration can thus be expected to enhance the clinical efficacy of WT1 immunotherapy.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cancer Vaccines/immunology , Interferon-beta/administration & dosage , Kidney Neoplasms/prevention & control , WT1 Proteins/immunology , Wilms Tumor/prevention & control , Animals , Cancer Vaccines/administration & dosage , Kidney Neoplasms/immunology , Killer Cells, Natural/immunology , Male , Mice , Mice, Inbred C57BL , Spleen/immunology , Survival Analysis , T-Lymphocytes, Cytotoxic/immunology , WT1 Proteins/administration & dosage , Wilms Tumor/immunology
17.
Anticancer Res ; 30(6): 2247-54, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20651376

ABSTRACT

Clinical studies of WT1-targeted cancer vaccine are being performed. However, WT1-specific Th response in cancer patients remains unclear. Using quantitative real-time RT-PCR, we investigated IFN-gamma and IL-10 mRNA expression from Th cells by stimulation with helper peptide WT1(332). Seventeen patients, of whom 10 had achieved stable disease and the remaining 7 had progressive disease, were weekly vaccinated with WT1 CTL epitope (modified WT1(235)) and examined for WT1(332)-specific Th response. A clear correlation between WT1(332)-specific Th response and clinical response was observed at 4 weeks post-vaccination. In patients who responded, a clear inverse correlation between IL-10-type and IFN-gamma-type WT1(332)-specific Th response was detected at pre- and 4 weeks post-vaccination, and the shift of the Th response from IL-10-type dominancy at early phase to IFN-gamma-type dominancy at late phase was observed. From this study we concluded that occurrence of WT1(332)-specific Th response could predict good clinical response of WT1 CTL epitope vaccination.


Subject(s)
Cancer Vaccines/immunology , Epitopes, T-Lymphocyte , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Vaccination , WT1 Proteins/immunology , Adult , Aged , Female , Humans , Interferon-gamma/genetics , Interleukin-10/genetics , Male , Middle Aged , RNA, Messenger/analysis
18.
Langmuir ; 25(19): 11221-4, 2009 Oct 06.
Article in English | MEDLINE | ID: mdl-19788204

ABSTRACT

SBA-15 thin films were synthesized by dip-coating in two different types of microtrenches: (a) silicon microtrenches and (b) silicon microtrenches with a deposited low-temperature oxide (LTO) layer. In the upper part of the synthesized films, the pores were aligned along the concave surface in both microtrenches. This alignment was attributed to the capillary force acting during solvent evaporation. In the lower part of the films, the pores were aligned tangentially with the wall in a silicon microtrench whereas they were aligned normal to the wall in a silicon microtrench with a deposited LTO layer. The LTO layer could suppress the growth of mesostructures from the substrate and promote growth from the vapor-liquid interface. The effects of the composition of the precursor solution and relative humidity on pore alignment were also clarified.

19.
Int J Oncol ; 34(5): 1181-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19360331

ABSTRACT

A high level protein synthesis is one of the characteristics of cancer cells. The aim of this study is to show the contribution of eukaryotic elongation factor 2 (eEF2), which plays an essential role in the polypeptide chain elongation step, in the tumorigenesis of gastrointestinal cancers. In the present study, we demonstrated by using immunohistochemistry that eEF2 protein was overexpressed in 92.9% (13 of 14) of gastric and 91.7% (22 of 24) of colorectal cancers. No mutations were found in any of the exons of the eEF2 gene in six gastric and six colorectal cancers. Knockdown of eEF2 by eEF2-specific short-hairpin RNA (shEF2) inhibited cancer cell growth in two gastric cancer cell lines, AZ-521 and MKN28, and one colon cancer cell line, SW620. Flow cytometric analysis showed that knockdown of eEF2 induced G2/M arrest and resulted in inactivation of Akt and cdc2 (a G2/M regulator) and activation of eEF2 kinase (a negative regulator of eEF2) in these cancer cells. Conversely, forced expression of eEF2 in AZ-521 cells significantly enhanced the cell growth through promotion of G2/M progression in cell cycle, activated Akt and cdc2, and inactivated eEF2 kinase. Furthermore, forced expression of eEF2 in these cancer cells enhanced in vivo tumorigenicity in a mouse xenograft model. These results showed that overexpressed eEF2 in gastrointestinal cancers promoted G2/M progression and enhanced their cell growth in vitro and in vivo. These results also suggested a novel linkage between translational elongation and cell cycle mechanisms, implying that the linkage might play an important role to orchestrate the deregulated translation and cell cycle mechanisms for promotion of the development of gastrointestinal cancers.


Subject(s)
Adenocarcinoma/genetics , Cell Division/genetics , G2 Phase/genetics , Gastrointestinal Neoplasms/genetics , Peptide Elongation Factor 2/genetics , Adult , Aged , Aged, 80 and over , Animals , Cell Cycle/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Models, Biological , Tumor Cells, Cultured , Up-Regulation , Young Adult
20.
Int J Cancer ; 125(2): 381-7, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19384943

ABSTRACT

There are urgent needs to develop methods for early detection of nonsmall cell lung cancer (NSCLC) because of its increasing incidence and poor prognosis. Here, we analyzed the production of IgG antibody (WT1 Ab) against WT1 (Wilms' tumor gene) protein that was overexpressed in the majority of NSCLC. Enzyme-linked immuno-sorbent assay showed that WT1 Ab was produced in all of 91 NSCLC patients and 70 healthy individuals and that WT1 Ab titers were significantly higher in NSCLC patients compared with healthy individuals. When the cut-off level of WT1 Ab titers were fixed at mean + 3SD of those in healthy individuals, 26.4% of NSCLC patients had WT1 Ab titers over the cut-off level, and positive rates of WT1 Ab at each clinical stage were 25.0, 30.8 and 38.4% in stage I, II and III NSCLC, respectively. When WT1 Ab was combined with CEA or CYFRA for detection of NSCLC, positive detection rates increased from 25.0 to 34.1 and 31.8%, respectively, in stage I and from 38.4 to 69.2 and 46.1%, respectively, in stage III, but not changed in stage II. Western blot analysis showed that dominant subclass of WT1 Ab was Th1-type IgG2. Interestingly, elevation of WT1 Ab titers was significantly associated with longer disease-free survival in patients with stages I-III NSCLC. These results showed that WT1 Ab could be a useful marker for early detection of NSCLC and its prognostic prediction. These results also suggested that WT1-specific immune responses played an important role in anti-cancer immunity in NSCLC.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Non-Small-Cell Lung/diagnosis , Immunoglobulin G/blood , Lung Neoplasms/diagnosis , WT1 Proteins/genetics , Blotting, Western , Carcinoma, Non-Small-Cell Lung/immunology , Early Diagnosis , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Lung Neoplasms/immunology , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL
...