Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Curr Issues Mol Biol ; 44(9): 3809-3821, 2022 Aug 24.
Article in English | MEDLINE | ID: mdl-36135173

ABSTRACT

The newly synthesized compound TGF-ß signaling agonist (T74) is a small molecule associated with the TGF-ß receptor signaling pathway. Tolerogenic dendritic cells (tDCs) have been used to examine immunosuppressive and anti-inflammatory effects in multiple autoimmune disease models. The aim of this study was to investigate whether treatment of DCs with T74 has an antirheumatic effect in a mouse model of collagen-induced arthritis (CIA). Bone marrow-derived cells were obtained from DBA/1J mice and differentiated into DCs. T74-treated DCs (T74-DCs) were generated by treating bone marrow-derived DCs with LPS, type II collagen, and T74. T74-DCs expressed lower levels of surface molecules and inflammatory cytokines associated with antigen presentation and T cell stimulation. The ability of T74-DCs to differentiate effector T cells was lower than that of T74-untreated DCs (NT-DCs), but T74-DCs increased the regulatory T (Treg) cell differentiation in vitro. DBA/1J mice received two subcutaneous (s.c.) injections of type II collagen to establish CIA. Mice then received two s.c. injections of T74-DCs or NT-DCs. Joint inflammation was ameliorated in the paws of T74-DC-treated mice. Additionally, Treg populations in T74-DC-treated mice were higher than in NT-DC-treated or PBS-treated CIA mice. Taken together, these results demonstrate that T74 induces tolerance in DCs, and that T74-mediated DCs exert antirheumatic effects via induction of Tregs.

2.
Int J Mol Sci ; 23(10)2022 May 18.
Article in English | MEDLINE | ID: mdl-35628458

ABSTRACT

Mitophagy is a selective form of autophagy that removes damaged mitochondria. Increasing evidence indicates that dysregulated mitophagy is implicated in numerous autoimmune diseases, but the role of mitophagy in rheumatoid arthritis (RA) has not yet been reported. The aim of the present study was to determine the roles of mitophagy in patient-derived RA synovial fibroblasts (RASFs) and in the collagen antibody-induced arthritis mouse model. We measured the mitophagy marker PTEN-induced putative kinase 1 (PINK1) in RASFs treated with tumor necrosis factor-α (TNF-α) using Western blotting and immunofluorescence. Arthritis was induced in PINK1-/- mice by intraperitoneal injection of an anti-type II collagen antibody cocktail and lipopolysaccharide. RA severity was assessed by histopathology. PINK1 expression and damaged mitochondria increased in TNF-α treated RASFs via increased intracellular levels of reactive oxygen species. PINK1 knockdown RASFs decreased cellular migration and invasion functions. In addition, PINK1-/- mice with arthritis exhibited markedly reduced swelling and inflammation relative to wild-type mice with arthritis. Taken together, these findings suggest that regulation of PINK1 expression in RA could represent a potential therapeutic and diagnostic target for RA.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Synovitis , Animals , Antibodies , Fibroblasts/metabolism , Humans , Mice , Mice, Knockout , Mitophagy , Protein Kinases/genetics , Protein Kinases/metabolism , Tumor Necrosis Factor-alpha/pharmacology
3.
Biomedicines ; 9(11)2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34829956

ABSTRACT

Enpp2 is an enzyme that catalyzes the conversion of lysophosphatidylcholine (LPC) to lysophosphatidic acid (LPA), which exhibits a wide variety of biological functions. Here, we examined the biological effects of Enpp2 on dendritic cells (DCs), which are specialized antigen-presenting cells (APCs) characterized by their ability to migrate into secondary lymphoid organs and activate naïve T-cells. DCs were generated from bone marrow progenitors obtained from C57BL/6 mice. Enpp2 levels in DCs were regulated using small interfering (si)RNA or recombinant Enpp2. Expression of Enpp2 in LPS-stimulated mature (m)DCs was high, however, knocking down Enpp2 inhibited mDC function. In addition, the migratory capacity of mDCs increased after treatment with rmEnpp2; this phenomenon was mediated via the RhoA-mediated signaling pathway. Enpp2-treated mDCs showed a markedly increased capacity to migrate to lymph nodes in vivo. These findings strongly suggest that Enpp2 is necessary for mDC migration capacity, thereby increasing our understanding of DC biology. We postulate that regulating Enpp2 improves DC migration to lymph nodes, thus improving the effectiveness of cancer vaccines based on DC.

4.
Int J Mol Sci ; 22(16)2021 Aug 13.
Article in English | MEDLINE | ID: mdl-34445415

ABSTRACT

Chimeric antigen receptor (CAR)-T cells are effective in the treatment of hematologic malignancies but have shown limited efficacy against solid tumors. Here, we demonstrated an approach to inhibit recurrence of B cell lymphoma by co-expressing both a human anti-CD19-specific single-chain variable fragment (scFv) CAR (CD19 CAR) and a TGF-ß/IL-7 chimeric switch receptor (tTRII-I7R) in T cells (CD19 CAR-tTRII-I7R-T cells). The tTRII-I7R was designed to convert immunosuppressive TGF-ß signaling into immune-activating IL-7 signaling. The effect of TGF-ß on CD19 CAR-tTRII-I7R-T cells was assessed by western blotting. Target-specific killing by CD19 CAR-tTRII-I7R-T cells was evaluated by Eu-TDA assay. Daudi tumor-bearing NSG (NOD/SCID/IL2Rγ-/-) mice were treated with CD19 CAR-tTRII-I7R-T cells to analyze the in vivo anti-tumor effect. In vitro, CD19 CAR-tTRII-I7R-T cells had a lower level of phosphorylated SMAD2 and a higher level of target-specific cytotoxicity than controls in the presence of rhTGF-ß1. In the animal model, the overall survival and recurrence-free survival of mice that received CD19 CAR-tTRII-I7R-T cells were significantly longer than in control mice. These findings strongly suggest that CD19 CAR-tTRII-I7R-T cell therapy provides a new strategy for long-lasting, TGF-ß-resistant anti-tumor effects against B cell lymphoma, which may lead ultimately to increased clinical efficacy.


Subject(s)
Antigens, CD19/immunology , Interleukin-7/genetics , Lymphoma, B-Cell/therapy , Neoplasm Recurrence, Local/therapy , Single-Chain Antibodies/metabolism , Transforming Growth Factor beta/genetics , Animals , Cells, Cultured , Female , Humans , Immunotherapy, Adoptive , Interleukin-7/metabolism , K562 Cells , Lymphoma, B-Cell/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Recurrence, Local/immunology , Receptors, Chimeric Antigen/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Treatment Outcome , Xenograft Model Antitumor Assays
5.
Int J Mol Sci ; 22(9)2021 Apr 23.
Article in English | MEDLINE | ID: mdl-33922658

ABSTRACT

Dendritic cells (DCs) are the most potent professional antigen-presenting cells (APCs) and inducers of T cell-mediated immunity. Although DCs play a central role in promoting adaptive immune responses against growing tumors, they also establish and maintain peripheral tolerance. DC activity depends on the method of induction and/or the presence of immunosuppressive agents. Tolerogenic dendritic cells (tDCs) induce immune tolerance by activating CD4+CD25+Foxp3+ regulatory T (Treg) cells and/or by producing cytokines that inhibit T cell activation. These findings suggest that tDCs may be an effective treatment for autoimmune diseases, inflammatory diseases, and infertility.


Subject(s)
Autoimmune Diseases/pathology , Dendritic Cells/immunology , Immune Tolerance/immunology , Infertility/pathology , Inflammation/pathology , Animals , Autoimmune Diseases/immunology , Humans , Infertility/immunology , Inflammation/immunology
6.
J Cancer ; 11(4): 769-775, 2020.
Article in English | MEDLINE | ID: mdl-31949478

ABSTRACT

Dendritic cells (DCs) have a critical effect on the outcome of adaptive immune responses against growing tumors. Recent studies on the metabolism on DCs provide new insights on the functioning of these critical controllers of innate and adaptive immunity. DCs within the tumor microenvironment (TME) often exist in an inactive state, which is thought to limit the adaptive immune response elicited by the growing tumor. Tumor-derived factors in the TME are known to suppress DC activation and result in functional alterations in DC phenotype. We are now beginning to appreciate that many of these factors can also induce changes in immune cell metabolism. In this review, we discuss the functional alternation of DC phenotype by tumor metabolites.

7.
Cancer Lett ; 469: 162-172, 2020 01 28.
Article in English | MEDLINE | ID: mdl-31634527

ABSTRACT

Treatment with chimeric antigen receptor (CAR)-modified T cells targeting CD19 has proved successful in patients with relapsed/refractory B cell malignancies. However, long-term follow-up indicates that remission in a substantial proportion of patients is not sustainable. Most patients that experience recurrence have tumors and lost the CAR-T cells. To maintain the activity of CAR-T cells, Raji-B-NDG mice were treated sequentially with CAR-T-19 cells and homologous cells expressing human CD19 to promote expansion of CAR-T cells. Sequential treatment of mice with CAR-T-19 cells followed by Raji tumor cells led to marked prolongation of survival. The best case scenario after sequential treatment was a survival time of more than 200 days; the average survival time of mice in the non-sequential treatment group was 80 days. We treated mice with autologous CD19-modified T cells after initial treatment with CAR-T-19 cells. The overall survival and recurrence-free survival times of mice receiving sequential treatment were significantly longer. The percentages of CAR+ T cells in peripheral blood increased. Sequential therapy with autologous CAR-T-19 and aT19 cells provides a new strategy for generating memory CAR-T cells, which may lead ultimately to increased clinical efficacy.


Subject(s)
Antigens, CD19/immunology , Immunotherapy, Adoptive/methods , Lymphoma, B-Cell/therapy , Neoplasm Recurrence, Local/prevention & control , T-Lymphocytes/transplantation , Animals , Antigens, CD19/genetics , Cell Line, Tumor , Combined Modality Therapy/methods , Disease-Free Survival , HEK293 Cells , Healthy Volunteers , Humans , Immunologic Memory , Longevity/immunology , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/mortality , Mice , Neoplasm Recurrence, Local/immunology , Receptors, Chimeric Antigen/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Remission Induction/methods , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Time Factors , Transduction, Genetic , Transplantation, Autologous/methods , Xenograft Model Antitumor Assays
8.
FASEB J ; 33(10): 11035-11044, 2019 10.
Article in English | MEDLINE | ID: mdl-31287961

ABSTRACT

Dendritic cells (DCs) are the most potent professional antigen (Ag)-presenting cells and inducers of T cell-mediated immunity. A previous microarray analysis identified PDZ and LIM domain protein 4 (Pdlim4) as a candidate marker for DC maturation. The aim of this study was to investigate whether Pdlim4 influences DC migration and maturation. Mouse bone marrow-derived DCs were transduced lentivirally with Pdlim4 short hairpin RNA and examined by confocal microscopy, flow cytometry, ELISA, and Western blotting. Pdlim4 was highly induced in LPS-stimulated mature DCs (mDCs). Pdlim4-knockdown mDCs showed reduced expression of molecules associated with Ag presentation and T-cell costimulation, reduced cytokine production, and functional defects in their ability to activate T cells. Moreover, Pdlim4 was necessary for mDC migration via C-C chemokine receptor type 7 (CCR7)-JNK in in vitro Transwell assays. The importance of Pdlim4 in DC migration was confirmed with an in vivo migration model in which C57BL/6 mice were injected with fluorescently labeled DCs in the footpad and migration to the popliteal lymph nodes was assessed by flow cytometry. Moreover, dendrite formation in mDCs was remarkably attenuated under Pdlim4 knockdown. Taken together, these results demonstrate that Pdlim4 is necessary for DC migration via CCR7-JNK, dendrite formation, and subsequent development of functional T-cell responses.-Yoo, J.-Y., Jung, N.-C., Lee, J.-H., Choi, S.-Y., Choi, H.-J., Park, S.-Y., Jang, J.-S., Byun, S.-H., Hwang, S.-U., Noh, K.-E., Park, Y., Lee, J., Song, J.-Y., Seo, H. G., Lee, H. S., Lim, D.-S. Pdlim4 is essential for CCR7-JNK-mediated dendritic cell migration and F-actin-related dendrite formation.


Subject(s)
Cell Movement , Dendritic Cells/metabolism , LIM Domain Proteins/metabolism , Microfilament Proteins/metabolism , Actins/metabolism , Animals , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/physiology , LIM Domain Proteins/genetics , Lymphocyte Activation , MAP Kinase Kinase 4/metabolism , Male , Mice , Mice, Inbred C57BL , Microfilament Proteins/genetics , Receptors, CCR7/metabolism
9.
Cell Death Dis ; 9(8): 823, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30068989

ABSTRACT

Dendritic cells (DCs) are the most potent professional antigen presenting cells and inducers of T cell-mediated immunity. However, few specific markers of mature DCs (mDC) have been reported. A previous microarray analysis revealed expression of mDC-specific genes and identified Rsad2 (radical S-adenosyl methionine domain containing 2) as a candidate specific marker for DC maturation. Mouse bone marrow-derived DCs were transfected with Rsad2 siRNA and examined by flow cytometry, ELISA, western, and confocal microscopy. C57BL/6 mice received intravenously B16F10 cells to establish a pulmonary metastasis model. Tumor-bearing mice then received subcutaneously two injections of mDCs or Rsad2 knockdown DCs. The cytotoxic T lymphocyte (CTL) population was examined from splenocytes of DC-vaccinated mice by flow cytometry. Rsad2 was induced at high levels in LPS-stimulated mDCs and mDC function was markedly attenuated under conditions of Rsad2 knockdown. Moreover, Rsad2 was necessary for mDC maturation via the IRF7-mediated signaling pathway. The importance of Rsad2 was confirmed in an Rsad2 knockdown lung metastasis mouse model in which mDCs lost their antitumor efficacy. Data on the CTL population further supported the results as above. Taken together, Rsad2 was an obvious and specific marker necessary for DC maturation and these findings will be clearly helpful for further understanding of DC biology.


Subject(s)
Dendritic Cells/immunology , Interferon Regulatory Factor-7/metabolism , Proteins/metabolism , Animals , Bone Marrow Cells/cytology , Cell Line, Tumor , Dendritic Cells/cytology , Dendritic Cells/drug effects , Interferon Type I/metabolism , Lipopolysaccharides/pharmacology , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Male , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Proteins/antagonists & inhibitors , Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology
10.
Transl Oncol ; 11(3): 686-690, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29627706

ABSTRACT

As a treatment for solid tumors, dendritic cell (DC)-based immunotherapy has not been as effective as expected. Here, we review the reasons underlying the limitations of DC-based immunotherapy for solid tumors and ask what can be done to improve immune cell-based cancer therapies. Several reports show that, rather than a lack of immune induction, the limited efficacy of DC-based immunotherapy in cases of renal cell carcinoma (RCC) likely results from inhibition of immune responses by tumor-secreted TGF-ß and an increase in the number of regulatory T (Treg) cells in and around the solid tumor. Indeed, unlike DC therapy for solid tumors, cytotoxic T lymphocyte (CTL) responses induced by DC therapy inhibit tumor recurrence after surgery; CTL responses also limit tumor metastasis induced by additional tumor-challenge in RCC tumor-bearing mice. Here, we discuss the mechanisms underlying the poor efficacy of DC-based therapy for solid tumors and stress the need for new and improved DC immunotherapies and/or combination therapies with killer cells to treat resistant solid tumors.

11.
Circulation ; 135(15): 1444-1457, 2017 Apr 11.
Article in English | MEDLINE | ID: mdl-28174192

ABSTRACT

BACKGROUND: Inflammatory responses play a critical role in left ventricular remodeling after myocardial infarction (MI). Tolerogenic dendritic cells (tDCs) can modulate immune responses, inducing regulatory T cells in a number of inflammatory diseases. METHODS: We generated tDCs by treating bone marrow-derived dendritic cells with tumor necrosis factor-α and cardiac lysate from MI mice. We injected MI mice, induced by a ligation of the left anterior descending coronary artery in C57BL/6 mice, twice with tDCs within 24 hours and at 7 days after the ligation. RESULTS: In vivo cardiac magnetic resonance imaging and ex vivo histology confirmed the beneficial effect on postinfarct left ventricular remodeling in MI mice treated with tDCs. Subcutaneously administered infarct lysate-primed tDCs near the inguinal lymph node migrated to the regional lymph node and induced infarct tissue-specific regulatory T-cell populations in the inguinal and mediastinal lymph nodes, spleen, and infarcted myocardium, indicating that a local injection of tDCs induces a systemic activation of MI-specific regulatory T cells. These events elicited an inflammatory-to-reparative macrophage shift. The altered immune environment in the infarcted heart resulted in a better wound remodeling, preserved left ventricular systolic function after myocardial tissue damage, and improved survival. CONCLUSIONS: This study showed that tDC therapy in a preclinical model of MI was potentially translatable into an antiremodeling therapy for ischemic tissue repair.


Subject(s)
Dendritic Cells/immunology , Macrophages/immunology , Myocardial Infarction/diagnosis , Myocardial Infarction/immunology , T-Lymphocytes, Regulatory/immunology , Ventricular Function, Left , Ventricular Remodeling , Adoptive Transfer , Animals , Antigens/immunology , Biomarkers , Cell Movement , Cell- and Tissue-Based Therapy , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Disease Models, Animal , Immunization , Lymphocyte Activation , Macrophages/metabolism , Magnetic Resonance Imaging , Male , Mice , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , Myocardium/immunology , Myocardium/pathology , Neovascularization, Pathologic , T-Lymphocytes, Regulatory/metabolism
12.
Immunol Invest ; 45(6): 553-65, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27410037

ABSTRACT

BACKGROUND: The response of hepatocellular carcinoma (HCC) to immunotherapy is often disappointing and new strategies are clearly needed. The aim of the present study was to investigate whether cytokine-induced killer (CIK) cells combined with a dendritic cell vaccination enhanced cytotoxicity against hepatocarcinoma tumor cells in an in vivo animal model. METHODS: CIKs and DCs were prepared from C3H/HeJ mice by conventional methods, the dendritic cell (DC) pulsed with a MH134 cell lysate, DC or CIK alone were used as controls. Cell phenotypes were analyzed by flow cytometry, cytokine secretion levels were determined by enzyme-linked immunosorbent assay (ELISA), and cytotoxicity was assessed by means of an in vitro lactate dehydrogenase (LDH) release assay. A mouse hepatocarcinoma cell MH134-bearing mice model was established to test the in vivo anti-tumor efficacy of the system. RESULTS: CIK cells combined with DC therapy resulted in significant inhibition of tumor growth compared with the control group, whereas the decrease in tumor growth in mice that had been treated with CIK or DC alone did not reach the level of statistical significance. The combination therapy led to a further increase in the population of cytotoxic T cells (CTLs) in vivo, compared to the CIK or DC alone therapy. In addition, the combination therapy significantly enhanced cytotoxic activity against MH134 cells. CONCLUSION: Taken together, these results show that a DC + CIK vaccination is more effective than DC or CIK alone therapy for the treatment of hepatocarcinoma cancer.


Subject(s)
Adoptive Transfer/methods , Carcinoma, Hepatocellular/therapy , Cytokine-Induced Killer Cells/transplantation , Dendritic Cells/transplantation , Liver Neoplasms/therapy , Animals , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , Cytokine-Induced Killer Cells/cytology , Cytokine-Induced Killer Cells/immunology , Cytokines/immunology , Cytokines/pharmacology , Cytotoxicity, Immunologic , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Humans , L-Lactate Dehydrogenase/metabolism , Liver Neoplasms/immunology , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C3H , Primary Cell Culture , Survival Analysis , T-Lymphocytes, Cytotoxic , Treatment Outcome , Tumor Burden
13.
Biochem Pharmacol ; 115: 85-93, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27208887

ABSTRACT

Rosiglitazone is a selective ligand for peroxisome proliferator-activated receptor-gamma (PPAR-γ), which serves diverse biological functions. A number of autoimmune disease models have been used to examine the anti-inflammatory and immunosuppressive effects of tolerogenic dendritic cells (tDCs). The aim of the present study was to investigate whether rosiglitazone-mediated DC (Rosi-DC) therapy suppressed arthritis in a collagen-induced arthritis (CIA) mouse model. Rosi-DCs were generated by treating immature DCs with TNF-α, type II collagen, and rosiglitazone. CIA mice then received subcutaneously (s.c.) two injections of Rosi-DCs. The severity of arthritis was then assessed histopathologically. The phenotypes of the DC and regulatory T (Treg) cell populations in CIA mice were determined by flow cytometry and the effect of Rosi-DCs on the secretion of autoimmunity-inducing cytokines was examined by ELISA. Rosi-DCs expressed lower levels of DC-related surface markers than mature DCs. Histopathological examination revealed that the degree of inflammation in the paws of Rosi-DC-treated mice was much lower than that in the paws of PBS-treated CIA mice. Taken together, these results clearly show that rosiglitazone-mediated DCs ameliorate CIA, most likely via the induction of antigen-specific Treg cells.


Subject(s)
Arthritis, Experimental/therapy , Dendritic Cells/drug effects , Dendritic Cells/transplantation , Thiazolidinediones/therapeutic use , Animals , Arthritis, Experimental/chemically induced , Arthritis, Experimental/immunology , Cells, Cultured , Coculture Techniques , Collagen , Dendritic Cells/immunology , Female , Mice , Mice, Inbred DBA , Rosiglitazone , T-Lymphocytes, Regulatory/immunology
14.
Autoimmunity ; 49(2): 90-101, 2016.
Article in English | MEDLINE | ID: mdl-26699759

ABSTRACT

Tolerogenic dendritic cells (tDCs) play an important role in inducing peripheral tolerance; however, few tDC-specific markers have been identified. The aims of this study were to examine whether tDCs show a different gene expression profile from that of immunogenic DCs and identify specific gene markers of each cell type, in DBA/1 mice. tDCs were generated by treating immature DCs (imDCs) with TNF-α and type II collagen. The gene expression profiles of mature (m)DCs and tDCs were then investigated by microarray analysis and candidate markers were validated by quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR). Supervised selection identified 75 gene signatures, 63 of which were consistently upregulated in mDCs and 12 of which were upregulated only in tDCs. Additionally, 10 genes were overexpressed or equally expressed in both tDCs and mDCs. Scin (tDC-specific genes) and Orm1, Pdlim4 and Enpp2 (mDC-specific genes) were validated by real-time qRT-PCR. Taken together, these results clearly show that tDCs and mDCs can be identified according to their expression of specific gene markers.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Regulation , Immune Tolerance/genetics , Transcriptome , Animals , Biomarkers , Cells, Cultured , Cluster Analysis , Coculture Techniques , Computational Biology/methods , Gene Expression Profiling , Gene Ontology , Immunophenotyping , Mice , Mice, Inbred DBA , Reproducibility of Results , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
15.
Cardiovasc Res ; 101(2): 203-10, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24189626

ABSTRACT

AIMS: Autoimmunity plays an important role in the pathogenesis of viral myocarditis and giant cell myocarditis. Experimental autoimmune myocarditis (EAM) is a mouse model of myocarditis that is induced by cardiac myosin. Tolerogenic dendritic cells (tDCs) are used as anti-inflammatory and immunosuppressive targets in a number of autoimmune disease models, but their effect on EAM has not been addressed. The aim of this study was to investigate whether tDC therapy in an EAM mouse model can suppress inflammatory myocarditis, a potential precursor of dilated cardiomyopathy. METHODS AND RESULTS: tDCs were generated by treating immature DCs (imDCs) with TNF-α and cardiac myosin. Mice with EAM were injected twice with tDCs (with a 1-week interval) at three doses (2 × 10(5), 1 × 10(6), or 2 × 10(6)). The severity of myocarditis was histopathologically assessed. The phenotypes of the DC and regulatory T (Treg) cell populations were determined by flow cytometry and the effect of tDCs on autoimmunity-inducing cytokines was examined by ELISA. Myosin-pulsed tDCs displayed lower levels of DC-related surface markers and expressed higher levels of indoleamine 2, 3-dioxygenase (IDO) than mature DCs (mDCs). Histopathological examination revealed that hearts from tDC-treated mice showed markedly reduced myocardial inflammation compared with those of untreated EAM mice. These therapeutic effects by tDCs were mediated at least by enhanced myosin-specific Treg cell induction and anti-inflammatory cytokine secretion. CONCLUSION: Taken together, these results show for the first time that myosin-pulsed tDCs ameliorate EAM, and that this occurs most likely via the induction of antigen-specific Treg cells.


Subject(s)
Adoptive Transfer , Autoimmune Diseases/prevention & control , Autoimmunity , Cardiac Myosins/immunology , Dendritic Cells/transplantation , Immune Tolerance , Myocarditis/prevention & control , Myocardium/immunology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/pathology , Cells, Cultured , Coculture Techniques , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Models, Animal , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Inflammation Mediators/metabolism , Male , Mice , Mice, Inbred BALB C , Myocarditis/immunology , Myocarditis/metabolism , Myocarditis/pathology , Myocardium/metabolism , Myocardium/pathology , Phenotype , T-Lymphocytes, Regulatory/immunology , Time Factors , Vaccination
16.
Int J Oncol ; 41(5): 1601-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22971679

ABSTRACT

Dendritic cells (DCs) are increasingly used as adjuvants for vaccination strategies; however, there has been very little development in DC vaccines for patients with hepatocellular carcinoma (HCC). In this study, we assessed the safety, feasibility and efficacy of a multiple tumor-associated antigen (TAA)-pulsed DC vaccine in 5 patients with advanced HCC. DCs were generated by culturing blood monocytes in the presence of granulocyte macrophage-colony stimulating factor and interleukin-4 for 5 days. The DC vaccine was prepared by pulsing DCs with cytoplasmic transduction peptide-attached α-fetoprotein, glypican-3 and MAGE-1 recombinant fusion proteins and cultivating them in the presence of maturation cocktail. DCs were injected subcutaneously near the inguinal lymph nodes, followed by topical application of toll-like receptor-7 agonist around the injection site. We showed that our DC vaccine was safe and well-tolerated over 6 vaccinations in 5 patients. All 5 patients showed T cell responses against TAAs. Clinical benefit was observed in one of the 5 patients. In conclusion, the feasibility, safety and immune activity of DCs pulsed with TAAs were confirmed in HCC patients. However, clinical response was detected only in one patient. Future trials may consider applying this therapy in a less advanced stage to obtain better clinical responses.


Subject(s)
Antigens, Neoplasm/immunology , Cancer Vaccines/therapeutic use , Carcinoma, Hepatocellular/therapy , Dendritic Cells/immunology , Liver Neoplasms/therapy , Aged , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Carcinoma, Hepatocellular/pathology , Cytokines/biosynthesis , Cytokines/immunology , Female , Humans , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , T-Lymphocytes/immunology , Treatment Outcome
17.
Cancer Lett ; 324(1): 58-65, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22554711

ABSTRACT

Dendritic cell (DC)-mediated immunotherapy has not been as effective as expected for most solid tumors. This study demonstrates immune responses against solid tumors mediated by DCs charged with photodynamic therapy (PDT)-induced tumor lysates, which contain the heat shock protein (HSP) 70. PDT tumor lysate-pulsed DC (PDT-DC) inhibited the growth of mammary EMT6 tumors to a greater extent than freeze/thawed tumor lysate-pulsed DC (FT-DC) or PDT tumor lysates. PDT-DC also showed significant anti-tumor effects against fully-established (i.e., late-stage) solid tumors. Taken together, these results suggest that PDT-DC vaccination is more effective than conventional DC therapy for the treatment of cancers.


Subject(s)
Dendritic Cells/immunology , Immunotherapy/methods , Neoplasms/drug therapy , Photochemotherapy/methods , Animals , Cancer Vaccines/immunology , Female , Freezing , HSP70 Heat-Shock Proteins/metabolism , Interleukin-2/metabolism , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/immunology , Mammary Neoplasms, Animal/pathology , Mice , Mice, Inbred BALB C , Neoplasms/immunology , Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL