Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Allergy ; 71(9): 1274-83, 2016 09.
Article in English | MEDLINE | ID: mdl-26990894

ABSTRACT

BACKGROUND: Regulatory T cells (Treg) represent a promising target for novel treatment strategies in patients with inflammatory/allergic diseases. A soluble derivate of the Treg surface molecule glycoprotein A repetitions predominant (sGARP) has strong anti-inflammatory and regulatory effects on human cells in vitro as well as in vivo through de novo induction of peripheral Treg. The aim of this study was to investigate the immunomodulatory function of sGARP and its possible role as a new therapeutic option in allergic diseases using a humanized mouse model. METHODS: To analyze the therapeutic effects of sGARP, adult NOD/Scidγc(-/-) (NSG) mice received peripheral blood mononuclear cells (PBMC) derived from allergic patients with sensitization against birch allergen. Subsequently, allergic inflammation was induced in the presence of Treg alone or in combination with sGARP. RESULTS: In comparison with mice that received Treg alone, additional treatment with sGARP reduced airway hyperresponsiveness (AHR), influx of neutrophils and macrophages into the bronchoalveolar lavage (BAL), and human CD45(+) cells in the lungs. Furthermore, the numbers of mucus-producing goblet cells and inflammatory cell infiltrates were reduced. To elucidate whether the mechanism of action of sGARP involves the TGF-ß receptor pathway, mice additionally received anti-TGF-ß receptor II (TGF-ßRII) antibodies. Blocking the signaling of TGF-ß through TGF-ßRII abrogated the anti-inflammatory effects of sGARP, confirming its essential role in inhibiting the allergic inflammation. CONCLUSION: Induction of peripheral tolerance via sGARP is a promising potential approach to treat allergic airway diseases.


Subject(s)
Inflammation/etiology , Inflammation/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Respiratory Hypersensitivity/etiology , Respiratory Hypersensitivity/metabolism , Adult , Allergens/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Female , Humans , Immune Tolerance , Immunoglobulin E/blood , Immunoglobulin E/immunology , Inflammation/pathology , Lung/immunology , Lung/metabolism , Lung/pathology , Male , Mice , Middle Aged , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Receptors, Transforming Growth Factor beta/metabolism , Respiratory Hypersensitivity/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
2.
Ann Oncol ; 17(4): 563-70, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16418308

ABSTRACT

BACKGROUND: This randomized phase III trial was designed to demonstrate the superiority of autologous peptide-loaded dendritic cell (DC) vaccination over standard dacarbazine (DTIC) chemotherapy in stage IV melanoma patients. PATIENTS AND METHODS: DTIC 850 mg/m2 intravenously was applied in 4-week intervals. DC vaccines loaded with MHC class I and II-restricted peptides were applied subcutaneously at 2-week intervals for the first five vaccinations and every 4 weeks thereafter. The primary study end point was objective response (OR); secondary end points were toxicity, overall (OS) and progression-free survival (PFS). RESULTS: At the time of the first interim analysis 55 patients had been enrolled into the DTIC and 53 into the DC-arm (ITT). OR was low (DTIC: 5.5%, DC: 3.8%), but not significantly different in the two arms. The Data Safety & Monitoring Board recommended closure of the study. Unscheduled subset analyses revealed that patients with normal serum LDH and/or stage M1a/b survived longer in both arms than those with elevated serum LDH and/or stage M1c. Only in the DC-arm did those patients with (i) an initial unimpaired general health status (Karnofsky = 100) or (ii) an HLA-A2+/HLA-B44- haplotype survive significantly longer than patients with a Karnofsky index <100 (P = 0.007 versus P = 0.057 in the DTIC-arm) or other HLA haplotypes (P = 0.04 versus P = 0.57 in DTIC-treated patients). CONCLUSIONS: DC vaccination could not be demonstrated to be more effective than DTIC chemotherapy in stage IV melanoma patients. The observed association of overall performance status and HLA haplotype with overall survival for patients treated by DC vaccination should be tested in future trials employing DC vaccines.


Subject(s)
Cancer Vaccines/administration & dosage , Dacarbazine/therapeutic use , Dendritic Cells/transplantation , Melanoma/therapy , Peptides/administration & dosage , Humans , Melanoma/pathology , Neoplasm Metastasis
3.
Gene Ther ; 10(3): 243-50, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12571632

ABSTRACT

In recent years, vaccination strategies using antigen-presenting cells (APC) have been under investigation. Antigen delivery using genetic immunization through ex vivo transduction of dendritic cells (DC) is supposed to enhance the induction of antitumor responses in humans by activating a broad range of peptide-specific CD8+ T cells. In this study, we compared the potential of adenoviral (Ad)-transduced versus peptide-pulsed DC to induce melanoma-antigen (Ag)-specific T-cell responses in vitro. Whereas gp100-peptide-pulsed DC induced long-lasting specific CD8+ T-cell responses against single peptides, Ad-transduced DC induced broad and strong, specific immunity against various peptides of the gp100-Ag. Surprisingly, several restimulations led to decreasing gp100-specific and in parallel to increasing anti-adenoviral T-cell responses. Nevertheless, those anti-adenoviral T-cell responses provided an "adjuvant" effect by inducing an early release of high amounts of IL-2/IFN-gamma, therewith enhancing CTL induction in the initiation phase. Based on these data, we suggest a prime/boost vaccination strategy in melanoma patients--combining the use of Ad-DC and peptide-pulsed DC--to obtain efficient and long-term antitumor T-cell responses.


Subject(s)
Dendritic Cells/immunology , Epitopes/genetics , Genetic Therapy/methods , Melanoma/therapy , Membrane Glycoproteins/genetics , Neoplasm Proteins/genetics , Skin Neoplasms/therapy , Adenoviridae/genetics , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines , Cell Line , Genetic Vectors/administration & dosage , Humans , Interferon-gamma/immunology , Interleukin-2/immunology , Melanoma/immunology , Membrane Glycoproteins/administration & dosage , Neoplasm Proteins/administration & dosage , Skin Neoplasms/immunology , Transduction, Genetic/methods , gp100 Melanoma Antigen
4.
Trends Immunol ; 22(7): 394-400, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11429324

ABSTRACT

The induction of antigen-specific T-cell tolerance in the thymus and its maintenance in the periphery is crucial for the prevention of autoimmunity. As well as their stimulatory functions, there is growing evidence that dendritic cells, acting as professional antigen-presenting cells, also maintain and regulate T-cell tolerance in the periphery. This control function is exerted by certain maturation stages and subsets of different ontogeny, and can be influenced by immunomodulatory agents. What is the current state of knowledge of the "immunoregulatory" properties of dendritic cells and how might tolerance-inducing dendritic cells be relevant to therapeutic applications in humans?


Subject(s)
Clonal Anergy/immunology , Dendritic Cells/immunology , T-Lymphocytes/immunology , Animals , Humans , Interleukin-10/immunology , Lymphocytes/immunology
5.
Int J Cancer ; 93(2): 243-51, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11410873

ABSTRACT

Dendritic cells (DCs) elicit potent anti-tumoral T-cell responses in vitro and in vivo. However, different types of DC have yet to be compared for their capacity to induce anti-tumor responses in vivo at different developmental stages. Herein, we correlated the efficiencies of different types of monocyte-derived DC as vaccines on the resulting anti-tumor immune responses in vivo. Immature and mature DCs were separately pulsed with a peptide derived from tyrosinase, MelanA/MART-1 or MAGE-1 and a recall antigen. Both DC populations were injected every 2 weeks in different lymph nodes of the same patient. Immune responses were monitored before, during and after vaccination. Mature DCs induced increased recall antigen-specific CD4(+) T-cell responses in 7/8 patients, while immature DCs did so in only 3/8. Expansion of peptide-specific IFN-gamma-producing CD8(+) T cells was observed in 5/7 patients vaccinated with mature DCs but in only 1/7 using immature DCs. However, these functional data did not correlate with the tetramer staining. Herein, immature DCs also showed expansion of peptide-specific T cells. In 2/4 patients vaccinated with mature DCs, we observed induction of peptide-specific cytotoxic T cells, as monitored by chromium-release assays, whereas immature DCs failed to induce peptide-specific cytotoxic T cells in the same patients. Instead, FCS-cultured immature DCs induced FCS-specific IgE responses in 1 patient. Our data demonstrate that this novel vaccination protocol is an efficient approach to compare different immunization strategies within the same patient. Thus, our data define FCS-free cultured mature DCs as superior inducers of T-cell responses in melanoma patients.


Subject(s)
Adjuvants, Immunologic/administration & dosage , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Melanoma/immunology , Antigens, Neoplasm , Humans , Immunization , Interferon-gamma/immunology , Lymph Nodes/immunology , Melanoma-Specific Antigens , Neoplasm Proteins/immunology , Neoplasm Staging , Peptides/immunology , T-Lymphocytes, Cytotoxic/immunology
6.
J Exp Med ; 193(11): 1285-94, 2001 Jun 04.
Article in English | MEDLINE | ID: mdl-11390435

ABSTRACT

A subpopulation of peripheral human CD4(+)CD25(+) T cells that expresses CD45RO, histocompatibility leukocyte antigen DR, and intracellular cytotoxic T lymphocyte-associated antigen (CTLA) 4 does not expand after stimulation and markedly suppresses the expansion of conventional T cells in a contact-dependent manner. After activation, CD4(+)CD25(+) T cells express CTLA-4 on the surface detectable for several weeks. These cells show a G1/G0 cell cycle arrest and no production of interleukin (IL)-2, IL-4, or interferon (IFN)-gamma on either protein or mRNA levels. The anergic state of CD4(+)CD25(+) T cells is not reversible by the addition of anti-CD28, anti-CTLA-4, anti-transforming growth factor beta, or anti-IL-10 antibody. However, the refractory state of CD4(+)CD25(+) T cells was partially reversible by the addition of IL-2 or IL-4. These data demonstrate that human blood contains a resident T cell population with potent regulatory properties.


Subject(s)
CD4 Antigens/analysis , Immunoconjugates , Receptors, Interleukin-2/analysis , T-Lymphocyte Subsets/physiology , Abatacept , Animals , Antigens, CD , Antigens, Differentiation/analysis , CTLA-4 Antigen , Cell Communication , Cytokines/biosynthesis , Humans , Immune Tolerance , Leukocyte Common Antigens/analysis , Lymphocyte Activation , Mice
7.
J Exp Med ; 192(9): 1213-22, 2000 Nov 06.
Article in English | MEDLINE | ID: mdl-11067871

ABSTRACT

The functional properties of dendritic cells (DCs) are strictly dependent on their maturational state. To analyze the influence of the maturational state of DCs on priming and differentiation of T cells, immature CD83(-) and mature CD83(+) human DCs were used for stimulation of naive, allogeneic CD4(+) T cells. Repetitive stimulation with mature DCs resulted in a strong expansion of alloreactive T cells and the exclusive development of T helper type 1 (Th1) cells. In contrast, after repetitive stimulation with immature DCs the alloreactive T cells showed an irreversibly inhibited proliferation that could not be restored by restimulation with mature DCs or peripheral blood mononuclear cells, or by the addition of interleukin (IL)-2. Only stimulation of T cells with mature DCs resulted in an upregulation of CD154, CD69, and CD70, whereas T cells activated with immature DCs showed an early upregulation of the negative regulator cytotoxic T lymphocyte-associated molecule 4 (CTLA-4). These T cells lost their ability to produce interferon gamma, IL-2, or IL-4 after several stimulations with immature DCs and differentiated into nonproliferating, IL-10-producing T cells. Furthermore, in coculture experiments these T cells inhibited the antigen-driven proliferation of Th1 cells in a contact- and dose-dependent, but antigen-nonspecific manner. These data show that immature and mature DCs induce different types of T cell responses: inflammatory Th1 cells are induced by mature DCs, and IL-10-producing T cell regulatory 1-like cells by immature DCs.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Dendritic Cells/immunology , Interleukin-10/metabolism , Lymphocyte Activation/immunology , Antigens/immunology , Antigens, CD/analysis , CD4-Positive T-Lymphocytes/cytology , Cell Differentiation , Cell Division , Dendritic Cells/cytology , Dose-Response Relationship, Immunologic , Flow Cytometry , Humans , Immunoglobulins/immunology , Immunophenotyping , Interleukin-2/immunology , Membrane Glycoproteins/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Transplantation, Homologous/immunology , CD83 Antigen
8.
J Invest Dermatol ; 115(4): 658-63, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10998139

ABSTRACT

Dendritic cells are key players of the immune system as they efficiently induce primary immune responses by activating naive T cells. We generated human dendritic cells from CD14+ blood precursors and investigated expression of the actin-bundling protein fascin during maturation by western blotting, immunofluorescence, and cytofluorometry. Cells obtained by culture of CD14+ blood precursors in the presence of granulocyte-macrophage colony-stimulating factor and interleukin-4, which were only weakly positive for the maturation marker CD83, expressed low amounts of fascin. Addition of a cytokine cocktail including tumor necrosis factor alpha, interleukin-1beta, interleukin-6, and prostaglandin E2 induced maturation of the cells and enhanced fascin expression in parallel with CD83 expression. Isolated mature CD83+ cells displayed especially high fascin levels on western blots, as did gated CD83+ dendritic cells in cytofluorometry. Dendritic cells generated from CD34+ blood precursors expressed high levels of fascin as well. Confocal microscopy revealed that location of fascin within the cell was restricted to the area of the submembranous actin cytoskeleton and to the dendritic processes. Suppression experiments using antisense constructs of fascin hint at a retarded morphologic maturation of dendritic cells, supporting the view that fascin expression is pivotal for dendrite formation. Our data suggest that fascin could serve as a marker molecule to monitor the maturation state of in vitro generated dendritic cells for use in clinical trials.


Subject(s)
Carrier Proteins/biosynthesis , Dendritic Cells/metabolism , Microfilament Proteins/biosynthesis , Actins/metabolism , Antigens, CD , Antigens, CD34/blood , Biomarkers/analysis , Blotting, Western , Cell Differentiation/drug effects , Cells, Cultured , Dendritic Cells/cytology , Humans , Immunoglobulins/analysis , Leukocytes, Mononuclear/immunology , Membrane Glycoproteins/analysis , Time Factors , CD83 Antigen
9.
J Immunol ; 165(6): 3492-6, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10975870

ABSTRACT

Dendritic cell (DC) vaccination, albeit still in an early stage, is a promising strategy to induce immunity to cancer. We explored whether DC can expand Ag-specific CD8+ T cells even in far-advanced stage IV melanoma patients. We found that three to five biweekly vaccinations of mature, monocyte-derived DC (three vaccinations of 6 x 106 s.c. followed by two i.v. ones of 6 and 12 x 106, respectively) pulsed with Mage-3A2.1 tumor and influenza matrix A2. 1-positive control peptides as well as the recall Ag tetanus toxoid (in three of eight patients) generated in all eight patients Ag-specific effector CD8+ T cells that were detectable in blood directly ex vivo. This is the first time that active, melanoma peptide-specific, IFN-gamma-producing, effector CD8+ T cells have been reliably observed in patients vaccinated with melanoma Ags. Therefore, our DC vaccination strategy performs an adjuvant role and encourages further optimization of this new immunization approach.


Subject(s)
Antigens, Neoplasm , Cancer Vaccines/immunology , Dendritic Cells/transplantation , Epitopes, T-Lymphocyte/immunology , Melanoma/immunology , Neoplasm Proteins/immunology , Peptides/immunology , T-Lymphocytes, Cytotoxic/immunology , Viral Matrix Proteins/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Cell Differentiation/immunology , Cells, Cultured , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/administration & dosage , Humans , Immunization, Secondary , Injections, Intravenous , Injections, Subcutaneous , Intercellular Signaling Peptides and Proteins , Lymphocyte Activation , Melanoma/pathology , Melanoma/therapy , Monocytes/immunology , Tetanus Toxoid/administration & dosage , Tetanus Toxoid/immunology
10.
Arch Dermatol Res ; 292(9): 437-45, 2000 Sep.
Article in English | MEDLINE | ID: mdl-11000287

ABSTRACT

Dendritic cells (DC) are the most potent antigen-presenting cells of the immune system. In this study we investigated the effects of various prostaglandins (PG) on the stimulatory capacity of DC. DC were generated from peripheral progenitor cells in the presence of IL-4 and GM-CSF and stimulated with IL-1, IL-6 and TNF-alpha on day 7. Simultaneously, PG (PGD(2), PGE(1), PGE(2), PGF(2 alpha), PGI(2)) were added at various concentrations (10(-5) to 10(-9) M) on day 7. In all experiments, PGE(2) had the most potent influence on the maturation of the DC, followed by other PG in the order PGE(1) > PGD(2) > PGF(2 alpha) > PGI(2). In addition, the expression of the surface molecules CD40, CD54, CD58, CD80, CD83, CD86 and the MHC class II molecules was upregulated after stimulation with PG. Analysis of DC supernatants after treatment with PG demonstrated significantly higher amounts of the proinflammatory cytokines IL-1 beta, IL-6, TNF-alpha, and IL-12. Addition of PG to DC induced a markedly enhanced proliferation of both naive and activated CD4(+) and CD8(+) T cells in alloantigen-induced MLR assays. Assessment of coculture supernatants after restimulation revealed significantly higher amounts of the Th1-cytokines IL-2 and IFN-gamma and only minimal amounts of IL-4 compared to control cells. No production of IL-10 was observed. The effects of PG on the maturation of DC and enhanced T-cell proliferation could be mimicked by db-cAMP and forskolin, indicating that they were due to elevated cAMP levels. Collectively, our data show that members of the PG family promote the differentiation of DC and enhance their capacity to induce a Th1 immune response.


Subject(s)
Cytokines/analysis , Dendritic Cells/immunology , Prostaglandins/pharmacology , Antigens, CD/analysis , Bucladesine , Cell Differentiation , Cells, Cultured , Coculture Techniques , Colforsin , Dendritic Cells/drug effects , Dose-Response Relationship, Drug , Humans , Interferon-gamma/analysis , Interleukin-1/analysis , Interleukin-12/analysis , Interleukin-2/analysis , Interleukin-6/analysis , Lymphocyte Activation , Prostaglandin D2/pharmacology , Tumor Necrosis Factor-alpha/analysis
11.
Cancer Res ; 60(16): 4446-52, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10969791

ABSTRACT

Dendritic cells (DCs) phagocytose apoptotic influenza-infected monocytes and cross-present influenza antigen to CD8+ T cells, generating a specific CTL response. We investigated whether apoptotic melanoma cells, presented by this mechanism, can lead to CTL responses to tumor-associated antigens and melanoma cells. Apoptotic HLA-A2- MEL-397 melanoma cells were internalized by HLA-A2+ immature monocyte-derived DCs but failed to induce maturation of DCs. When exposed to interleukin 6, interleukin 1beta, tumor necrosis factor alpha, and prostaglandin E2, DCs containing apoptotic MEL-397 cell material matured normally [cross-presenting DCs (cp-DCs)]. Autologous CD8+ CTL lines generated with cp-DCs produced tumor necrosis factor when stimulated with HLA-A2-binding immunodominant peptides from MelanA/MART1 and MAGE-3 (expressed by MEL-397 cells) but not tyrosinase (absent in MEL-397). T2 target cells loaded with the respective peptides were lysed by these cell lines, although to a lesser extent than by CTL lines generated in the presence of mature DCs and peptides from melanoma-associated h antigens. In contrast, lines generated with cp-DCs lysed HLA-A2+ MEL-526 melanoma cells or allogenic HLA-A2+ cp-DCs efficiently, whereas the CTL generated with DCs and peptides had little lytic activity. Mature DCs containing apoptotic tumor cells may thus represent an alternative approach for the therapy of malignant tumors.


Subject(s)
Antigen Presentation/immunology , Antigens, Neoplasm , Apoptosis/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Melanoma/immunology , Cytotoxicity, Immunologic/immunology , Epitopes, T-Lymphocyte/immunology , HLA-A2 Antigen/biosynthesis , HLA-A2 Antigen/immunology , Humans , Immunodominant Epitopes/immunology , Immunotherapy/methods , Lymphocyte Activation/immunology , MART-1 Antigen , Melanoma/pathology , Melanoma/therapy , Monophenol Monooxygenase/immunology , Neoplasm Proteins/immunology , Peptide Fragments/immunology , Phagocytosis , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
12.
Arch Dermatol Res ; 292(7): 325-32, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10966056

ABSTRACT

Tumor vaccination strategies using antigen-pulsed dendritic cells (DC) are currently under development. We established an in vitro system using cultured DC from HLA-typed volunteers for the induction of tumor peptide-specific CD8+ T cells. The strength and specificity of the resulting CTL responses were investigated. For stimulation of syngeneic CD8+ T cells two well-defined DC populations were generated: CD1a+ immature DC cultured in the presence of GM-CSF and IL-4 and mature CD83+ DC generated by additional stimulation with a cytokine cocktail. Stimulations were performed under serum-free conditions and in the absence of exogenous cytokines. Analysis of T cell responses showed that mature DC, but not immature DC, were able to induce the expansion of syngeneic tumor peptide-specific CD8+ T cells. Priming of CD8+ T cells with peptide-pulsed mature DC rapidly increased the frequency of antigen-specific T cells (ELISPOT technique). T cells induced by mature DC showed strong antigen-specific cytotoxicity in 51Cr-release assays whereas no antigen-specific cytotoxicity was detectable in CTL generated by immature DC. These data show that terminally differentiated mature DC are necessary for the induction of tumor antigen-specific CTL responses.


Subject(s)
Neoplasm Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Antigens, CD , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/physiology , Cell Differentiation/physiology , Cells, Cultured , Cellular Senescence , Culture Media, Serum-Free , Cytokines/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/physiology , Drug Combinations , Humans , Immunoglobulins/analysis , Membrane Glycoproteins/analysis , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/physiology , CD83 Antigen
13.
Gene Ther ; 7(3): 249-54, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10694802

ABSTRACT

We have developed a culture method for the foreign serum-free generation of highly immunostimulatory, CD83+ human dendritic cells (DC). In this study, we evaluated the feasibility and consequences of endogenously expressing antigens in mature DC using adenoviral vectors. Transduction of DC with Ad-EGFP demonstrated endogenous fluorescence in 50-85% of CD83+ DC. Ad-transduced DC stimulated the proliferation of allogeneic CD8+ and CD4+ T cells at low DC: T cell ratios. However, at high DC: T cell ratios the stimulatory capacity of Ad-transduced DC was suppressed. This immunosuppressive effect was confirmed by demonstrating that the stimulatory function of untreated DC could be suppressed in a dose-dependent manner by addition of Ad-transduced DC. Furthermore, transwell experiments suggested that direct cell contact was required. Taken together, our results demonstrate the feasibility of efficiently expressing antigens in CD83+ DC using adenoviruses. However, immunosuppressive effects must be considered and carefully studied before Ad-transduced DC are employed for clinical trials. Gene Therapy (2000) 7, 249-254.


Subject(s)
Adenoviridae/genetics , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/metabolism , Transduction, Genetic/genetics , CD4-CD8 Ratio , Flow Cytometry , Humans , Immunity, Active , Phenotype
14.
J Invest Dermatol ; 114(1): 142-9, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10620130

ABSTRACT

UNLABELLED: Standard protocols to generate mouse dendritic cells (DC) generally use culture medium supplemented with fetal calf serum; however, reinjection in vivo of DC cultured in fetal calf serum results in priming to xenogeneic proteins that clearly limits the use of such DC. We therefore established a fetal calf serum-free culture system for the generation of murine DC from bone marrow precursors. DC can be generated fetal calf serum-free using RPMI supplemented with 1.5% syngeneic mouse serum. Although the yield of DC grown under fetal calf serum-free conditions was somewhat lower than that of the standard culture, large numbers of DC could be generated without the exposure to xenogeneic proteins. The yield of fetal calf serum-free cultured DC was further enhanced by addition of the proinflammatory cytokines TNF-alpha and IL-1beta with the combination resulting in up to 10% more DC. Phenotypically, CD11c + DC cultured fetal calf serum-free homogenously coexpressed the DC-specific molecule DEC-205 as well as the costimulatory molecules CD40, CD80, and CD86. In contrast, only a subpopulation of the CD11c + DC cultured in fetal calf serum-containing medium coexpressed these molecules. Functionally, fetal calf serum-free DC showed strong stimulatory capacity for naïve allogeneic CD4 + and CD8 + T cells. Importantly, fetal calf serum-free DC showed spontaneous in vivo migratory activity. Moreover, 5 x 105 subcutaneously injected TNBS-conjugated fetal calf serum-free DC were able to mediate contact sensitivity. Furthermore, the intravenous or subcutaneous injection of a single dose of 5 x 105 OVA-pulsed fetal calf serum-free DC resulted in the induction of an OVA-specific immune response in naïve TCR transgenic animals. Thus DC cultured under fetal calf serum-free conditions are suitable instruments for in vivo therapeutic approaches, especially in autoimmune models. KEYWORDS: DC vaccines/dendritic cell development/fetal calf serum-free culture conditions for DC/in vivo therapeutic DC approaches.


Subject(s)
Dendritic Cells/cytology , Animals , Bone Marrow Cells/cytology , Cattle/embryology , Cell Count , Cell Division/drug effects , Cell Movement/physiology , Culture Media/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/physiology , Dermatitis, Contact/physiopathology , Fetal Blood/physiology , Interleukin-1/pharmacology , Mice , Mice, Inbred Strains , Mice, Transgenic/genetics , Ovalbumin/pharmacology , Phenotype , Receptors, Antigen, T-Cell/genetics , Stem Cells/cytology , Tumor Necrosis Factor-alpha/pharmacology
15.
J Exp Med ; 190(11): 1669-78, 1999 Dec 06.
Article in English | MEDLINE | ID: mdl-10587357

ABSTRACT

Dendritic cells (DCs) are considered to be promising adjuvants for inducing immunity to cancer. We used mature, monocyte-derived DCs to elicit resistance to malignant melanoma. The DCs were pulsed with Mage-3A1 tumor peptide and a recall antigen, tetanus toxoid or tuberculin. 11 far advanced stage IV melanoma patients, who were progressive despite standard chemotherapy, received five DC vaccinations at 14-d intervals. The first three vaccinations were administered into the skin, 3 x 10(6) DCs each subcutaneously and intradermally, followed by two intravenous injections of 6 x 10(6) and 12 x 10(6) DCs, respectively. Only minor (less than or equal to grade II) side effects were observed. Immunity to the recall antigen was boosted. Significant expansions of Mage-3A1-specific CD8(+) cytotoxic T lymphocyte (CTL) precursors were induced in 8/11 patients. Curiously, these immune responses often declined after the intravenous vaccinations. Regressions of individual metastases (skin, lymph node, lung, and liver) were evident in 6/11 patients. Resolution of skin metastases in two of the patients was accompanied by erythema and CD8(+) T cell infiltration, whereas nonregressing lesions lacked CD8(+) T cells as well as Mage-3 mRNA expression. This study proves the principle that DC "vaccines" can frequently expand tumor-specific CTLs and elicit regressions even in advanced cancer and, in addition, provides evidence for an active CD8(+) CTL-tumor cell interaction in situ as well as escape by lack of tumor antigen expression.


Subject(s)
Cancer Vaccines , Dendritic Cells/immunology , Dendritic Cells/transplantation , Lung Neoplasms/secondary , Melanoma/immunology , Melanoma/therapy , Neoplasm Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , Adult , Aged , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Female , Humans , Immunization Schedule , Lung Neoplasms/therapy , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Male , Melanoma/pathology , Middle Aged , Monocytes/immunology , Neoplasm Metastasis , Neoplasm Staging , Tetanus Toxoid/immunology , Tuberculin/immunology
16.
J Allergy Clin Immunol ; 104(5): 1052-9, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10550752

ABSTRACT

BACKGROUND: Atopy and IgE production are associated with enhanced allergen-specific T(H)2 responses. Therefore a causative treatment may result from the deviation of this T(H)2-dominated immune response toward a T(H)1 response. OBJECTIVE: This study was carried out to analyze whether dendritic cells, the most potent antigen-presenting cells that are also known to induce antigen-specific T(H)1 responses, are suitable for therapy of atopic diseases by shifting the allergen-specific T(H)2 response toward a T(H)1 response. METHODS: Monocyte-derived dendritic cells were used to present allergens in vitro to autologous CD4(+) T cells of allergic persons. Because collagen type I activates dendritic cells and enhances the secretion of IL-12, we performed allergen presentation assays also in the presence of collagen type I. RESULTS: After stimulation with allergen-pulsed dendritic cells the production of IFN-gamma as well as that of IL-4 and IL-5 by CD4(+) T cells was enhanced. In the presence of collagen type I, however, a significant shift toward a T(H)1 response with increased production of IFN-gamma and a decreased production of IL-5 could be observed. When T cells were stimulated directly with anti-CD3 and anti-CD28 in the absence of antigen-presenting cells, it was demonstrated that collagen type I also exerted a direct effect on T cells, increasing their IFN-gamma production. CONCLUSION: These data indicate that collagen type I influences dendritic cells as well as T cells in a way that a shift in cytokine production results in a T(H)1 response even in already-sensitized atopic individuals.


Subject(s)
Allergens/immunology , Collagen/immunology , Dendritic Cells/immunology , Hypersensitivity, Immediate/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Antigen Presentation/immunology , Cells, Cultured , Cytokines/biosynthesis , Humans , Interferon-gamma/biosynthesis , Interleukin-4/biosynthesis , Interleukin-5/biosynthesis , Lymphocyte Activation , Neutralization Tests , T-Lymphocytes/immunology
17.
Blood ; 93(5): 1634-42, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10029592

ABSTRACT

Dendritic cells (DC) are critically involved in the initiation of primary immune processes, including tumor rejection. In our study, we investigated the effect of interleukin-10 (IL-10)-treated human DC on the properties of CD8(+) T cells that are known to be essential for the destruction of tumor cells. We show that IL-10-pretreatment of DC not only reduces their allostimulatory capacity, but also induces a state of alloantigen-specific anergy in both primed and naive (CD45RA+) CD8(+) T cells. To investigate the influence of IL-10-treated DC on melanoma-associated antigen-specific T cells, we generated a tyrosinase-specific CD8(+) T-cell line by several rounds of stimulation with the specific antigen. After coculture with IL-10-treated DC, restimulation of the T-cell line with untreated, antigen-pulsed DC demonstrated peptide-specific anergy in the tyrosinase-specific T cells. Addition of IL-2 to the anergic T cells reversed the state of both alloantigen- or peptide-specific anergy. In contrast to optimally stimulated CD8(+) T cells, anergic tyrosinase-specific CD8(+) T cells, after coculture with peptide-pulsed IL-10-treated DC, failed to lyse an HLA-A2-positive and tyrosinase-expressing melanoma cell line. Thus, our data demonstrate that IL-10-treated DC induce an antigen-specific anergy in cytotoxic CD8(+) T cells, a process that might be a mechanism of tumors to inhibit immune surveillance by converting DC into tolerogenic antigen-presenting cells.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Interleukin-10/pharmacology , Lymphocyte Activation/drug effects , Melanoma/immunology , Skin Neoplasms/immunology , Antigen Presentation/drug effects , Antigens, Neoplasm/immunology , Cells, Cultured , Coculture Techniques , Cytotoxicity, Immunologic/drug effects , Dendritic Cells/pathology , Humans , Interleukin-10/immunology , Melanoma/pathology , Skin Neoplasms/pathology
18.
Eur J Immunol ; 28(10): 3231-9, 1998 10.
Article in English | MEDLINE | ID: mdl-9808192

ABSTRACT

IL-18 is a recently described cytokine that shares biological activities with IL-12 in driving the development of Th1-type T cells. As dendritic cells (DC) are very potent inducers of T cell proliferation and differentiation we wondered whether they utilize IL-18 as a factor driving Th1 development. We demonstrate by Northern blot and reverse transcription-PCR that various subtypes of human and murine DC as well as the DC-line XS contain IL-18 mRNA. When supernatants of either enriched Langerhans cells (LC) or bone marrow-derived DC were analyzed for production of IL-18 protein, IL-18 production was detected in an IL-18-specific ELISA. To assess whether the IL-18 protein released by DC is functional, we performed a sensitive bioassay using the IL-18-dependent stimulation of concanavalin A-stimulated T cells. Both, supernatants from bone marrow-derived DC and enriched LC induced IFN-gamma production in the T cells. This production was partially inhibitable by addition of anti-IL-18 antiserum. In a TCR-transgenic mouse system we further demonstrate that DC-derived IL-18 potentiates IL-12-dependent Th1 development. Using DC derived from IL-12 knockout animals, we show that DC-derived IL-18 by itself is not capable of inducing Th1 cell differentiation. Together the data demonstrate that subtypes of DC are able to release functional IL-18 that is able to induce IFN-gamma production and Th1 differentiation in primed T cells.


Subject(s)
Dendritic Cells/metabolism , Interleukin-12/metabolism , Interleukin-18/biosynthesis , Th1 Cells/metabolism , Animals , Blotting, Northern , Cell Differentiation , Cell Line , Culture Media , Humans , Interleukin-18/genetics , Langerhans Cells/metabolism , Mice , Mice, Transgenic , RNA, Messenger
19.
Eur J Immunol ; 28(5): 1673-80, 1998 May.
Article in English | MEDLINE | ID: mdl-9603474

ABSTRACT

The development of dendritic cells (DC) is still only partly understood. Recently established culture systems using CD34+ cells or monocytes as precursor cells for the generation of DC indicate the necessity of pro-inflammatory cytokines for their development. In vivo the contact to other cells or to the proteins of the extracellular matrix might also be essential for their development. In our experiments we used granulocyte-macrophage colony-stimulating factor- and IL-4-treated human monocytes as precursor cells to investigate the interaction of DC at different maturation stages with the matrix proteins fibronectin, collagen type I and collagen type IV. We demonstrate a strong beta1-integrin-mediated adherence of immature DC to fibronectin that is lost completely during maturation. The binding to collagen type I was less strong but induced a maturation of the precursor cells. After 3 days of culture on this protein, the cells showed all features of fully matured DC such as expression of CD83 and an excellent allostimulatory capacity. The reason for this effect was shown to be the induction of TNF-alpha production by the DC themselves. In contrast to the adhesion to fibronectin, the maturation and the cytokine production of DC induced by collagen type I could not be inhibited by blocking of beta1-integrins. These results indicate that proteins of the extracellular matrix play an important role in the development and function of human DC.


Subject(s)
Dendritic Cells/cytology , Extracellular Matrix Proteins/physiology , Antigens, Surface/biosynthesis , Antigens, Surface/drug effects , Cell Adhesion/immunology , Cell Communication/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Cells, Cultured , Collagen/physiology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Fibronectins/physiology , Humans , Integrin beta1/physiology , Lymphocyte Culture Test, Mixed , Monocytes/cytology , Monocytes/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Up-Regulation/immunology
20.
J Immunol ; 160(2): 667-73, 1998 Jan 15.
Article in English | MEDLINE | ID: mdl-9551901

ABSTRACT

To investigate the intracellular signaling mechanisms involved in the activation of APC by contact sensitizers, we studied the induction of tyrosine phosphorylation by these agents. Selective analysis of phosphotyrosine (p-tyr) in human Langerhans cells and different mononuclear cell types was achieved using a multicolor flow-cytometric technique. Stimulation with contact sensitizers revealed a distinct increase in p-tyr exclusively for MHC class II-positive cells. For different haptens, irritants, as well as activators of distinct signal transduction pathways, it was demonstrated that only strong sensitizers or the protein tyrosine phosphatase inhibitor sodium orthovanadate or cross-linking of MHC class II molecules were able to induce formation of p-tyr in human blood-derived dendritic cells serving as model for the dendritic cell family. This event required physiologic cell culture conditions and was blocked by specific inhibitors of protein tyrosine kinases. No evidence for the inhibition of protein tyrosine phosphatases by haptens was found. Western blot analysis of monocyte-enriched populations revealed an augmented phosphorylation of distinct proteins after hapten stimulation partly resembling the pattern noticed after cross-linking of HLA-DR molecules. In dendritic cells generated from mononuclear progenitors, the protein tyrosine kinase inhibitor genistein was able to block tyrosine phosphorylation as well as production of IL-1beta mRNA transcripts. Our data underline the unique capacity of haptens to activate APC and the important role of tyrosine phosphorylation for this process.


Subject(s)
Antigen-Presenting Cells/metabolism , Dermatitis, Contact/immunology , Haptens/pharmacology , Histocompatibility Antigens Class II/metabolism , Phosphotyrosine/metabolism , Cells, Cultured , Dendritic Cells/enzymology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dermatitis, Contact/metabolism , Dinitrofluorobenzene/pharmacology , Enzyme Activation/drug effects , Enzyme Activation/immunology , Genistein/pharmacology , Humans , Interleukin-1/antagonists & inhibitors , Interleukin-1/biosynthesis , Interleukin-1/genetics , Lymphocytes/metabolism , Monocytes/immunology , Monocytes/metabolism , Phosphorylation , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...