Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 May 03.
Article in English | MEDLINE | ID: mdl-38746459

ABSTRACT

NK cells are innate lymphocytes critical for surveillance of viruses and tumors, however the mechanisms underlying NK cell dysfunction in cancer are incompletely understood. We assessed the effector function of NK cells from bladder cancer patients and found severe dysfunction in NK cells derived from tumors versus peripheral blood. While both peripheral and tumor-infiltrating NK cells exhibited conserved patterns of inhibitory receptor over-expression, this did not explain the observed defects in NK surveillance in bladder tumors. Rather, TME-specific TGF-ß and metabolic perturbations such as hypoxia directly suppressed NK cell function. Specifically, an oxygen-dependent reduction in signaling through SLAMF6 was mechanistically responsible for poor NK cell function, as tumor-infiltrating NK cells cultured ex vivo under normoxic conditions exhibited complete restoration of function, while deletion of SLAMF6 abrogated NK cell cytolytic function even under normoxic conditions. Collectively, this work highlights the role of tissue-specific factors in dictating NK cell function, and implicates SLAMF6 signaling as a rational target for immuno-modulation to improve NK cell function in bladder cancer.

2.
Methods Mol Biol ; 2618: 133-145, 2023.
Article in English | MEDLINE | ID: mdl-36905514

ABSTRACT

Dendritic cells (DCs) represent one of the most important immune cell subsets in preventing the host from pathogen invasion by promoting both innate and adaptive immunity. Most research on human dendritic cells has focused on the easy-to-obtain dendritic cells derived in vitro from monocytes (MoDCs). However, many questions remain unanswered regarding the role of different dendritic cell types. The investigation of their roles in human immunity is hampered by their rarity and fragility, which especially holds true for type 1 conventional dendritic cells (cDC1s) and for plasmacytoid dendritic cells (pDCs). In vitro differentiation from hematopoietic progenitors emerged as a common way to produce different DC types, but the efficiency and reproducibility of these protocols needed to be improved and the extent to which the DCs generated in vitro resembled their in vivo counterparts required a more rigorous and global assessment. Here, we describe a cost-effective and robust in vitro differentiation system for the production of cDC1s and pDCs equivalent to their blood counterparts, from cord blood CD34+ hematopoietic stem cells (HSCs) cultured on a stromal feeder layer with a combination of cytokines and growth factors.


Subject(s)
Dendritic Cells , Hematopoietic Stem Cells , Humans , Reproducibility of Results , Cell Differentiation , Antigens, CD34/metabolism
3.
iScience ; 25(7): 104599, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35789859

ABSTRACT

Pattern recognition receptors (PRRs) protect against microbial invasion by detecting specific molecular patterns found in pathogens and initiating an immune response. Although microbial-derived PRR ligands have been extensively characterized, the contribution and relevance of endogenous ligands to PRR activation remains overlooked. Here, we characterize the landscape of endogenous ligands that engage RIG-I-like receptors (RLRs) upon infection by different RNA viruses. In each infection, several RNAs transcribed by RNA polymerase III (Pol3) specifically engaged RLRs, particularly the family of Y RNAs. Sensing of Y RNAs was dependent on their mimicking of viral secondary structure and their 5'-triphosphate extremity. Further, we found that HIV-1 triggered a VPR-dependent downregulation of RNA triphosphatase DUSP11 in vitro and in vivo, inducing a transcriptome-wide change of cellular RNA 5'-triphosphorylation that licenses Y RNA immunogenicity. Overall, our work uncovers the contribution of endogenous RNAs to antiviral immunity and demonstrates the importance of this pathway in HIV-1 infection.

4.
Adv Immunol ; 148: 49-91, 2020.
Article in English | MEDLINE | ID: mdl-33190733

ABSTRACT

Dendritic cells are a specialized subset of hematopoietic cells essential for mounting immunity against tumors and infectious disease as well as inducing tolerance for maintenance of homeostasis. DCs are equipped with number of immunoregulatory or stimulatory molecules that interact with other leukocytes to modulate their functions. Recent advances in DC biology identified a specific role for the conventional dendritic cell type 1 (cDC1) in eliciting cytotoxic CD8+ T cells essential for clearance of tumors and infected cells. The critical role of this subset in eliciting immune responses or inducing tolerance has largely been defined in mice whereas the biology of human cDC1 is poorly characterized owing to their extremely low frequency in tissues. A detailed characterization of the functions of many immunoregulatory and stimulatory molecules expressed by human cDC1 is critical for understanding their biology to exploit this subset for designing novel therapeutic modalities against cancer, infectious disease and autoimmune disorders.


Subject(s)
Autoimmune Diseases/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Infections/immunology , Neoplasms/immunology , Animals , Cytotoxicity, Immunologic , Humans , Immune Tolerance , Immunity, Cellular
5.
Cancer Cell ; 37(6): 751-753, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32516584

ABSTRACT

Migratory dendritic cells (DCs) in tumors transport antigens and share them with lymph node resident DCs through cross-presentation. In this issue of Cancer Cell, Ruhland et al. demonstrate that transport and transfer of tumor antigens in vesicles is a dominant pathway to load resident DCs for presentation to T cells.


Subject(s)
Antigens, Neoplasm , Cross-Priming , Antigens , Cell Movement , Dendritic Cells , Lymph Nodes
6.
Nat Cancer ; 1(12): 1204-1217, 2020 12.
Article in English | MEDLINE | ID: mdl-35121932

ABSTRACT

Generating responses to tumor antigens poses a challenge for immunotherapy. This phase II trial (NCT02129075) tested fms-like tyrosine kinase 3 (Flt3) ligand pre-treatment enhancement of responses to dendritic cell (DC)-targeting vaccines. We evaluated a regimen of Flt3L (CDX-301) to increase DCs and other antigen-presenting cells, poly-ICLC (TLR3 agonist that activates DCs) and a vaccine comprising anti-DEC-205-NY-ESO-1, a fusion antibody targeting CD205, linked to NY-ESO-1. High-risk melanoma patients were randomized to vaccine, with and without CDX-301. The end point was immune response to NY-ESO-1. Flt3L increased peripheral monocytes and conventional DCs (cDCs), including cross-presenting cDC1 and cDC2 and plasmacytoid DCs. Significant increases in humoral and T-cell responses and activation of DCs, natural killer cells and T cells were elicited. Transcriptional analyses revealed gene signatures associated with CDX-301 induction of an early, durable immune response. This study reveals in vivo effects of Flt3L on innate immune cells in the setting of vaccination, leading to an immunogenic vaccine regimen.


Subject(s)
Cancer Vaccines , Melanoma , Dendritic Cells , Humans , Immunity , Membrane Proteins , fms-Like Tyrosine Kinase 3
7.
Cancer Immunol Res ; 8(1): 70-80, 2020 01.
Article in English | MEDLINE | ID: mdl-31699709

ABSTRACT

Given its ability to induce both humoral and cellular immune responses, NY-ESO-1 has been considered a suitable antigen for a cancer vaccine. Despite promising results from early-phase clinical studies in patients with melanoma, NY-ESO-1 vaccine immunotherapy has not been widely investigated in larger trials; consequently, many questions remain as to the optimal vaccine formulation, predictive biomarkers, and sequencing and timing of vaccines in melanoma treatment. We conducted an adjuvant phase I/II clinical trial in high-risk resected melanoma to optimize the delivery of poly-ICLC, a TLR-3/MDA-5 agonist, as a component of vaccine formulation. A phase I dose-escalation part was undertaken to identify the MTD of poly-ICLC administered in combination with NY-ESO-1 and montanide. This was followed by a randomized phase II part investigating the MTD of poly-ICLC with NY-ESO-1 with or without montanide. The vaccine regimens were generally well tolerated, with no treatment-related grade 3/4 adverse events. Both regimens induced integrated NY-ESO-1-specific CD4+ T-cell and humoral responses. CD8+ T-cell responses were mainly detected in patients receiving montanide. T-cell avidity toward NY-ESO-1 peptides was higher in patients vaccinated with montanide. In conclusion, NY-ESO-1 protein in combination with poly-ICLC is safe, well tolerated, and capable of inducing integrated antibody and CD4+ T-cell responses in most patients. Combination with montanide enhances antigen-specific T-cell avidity and CD8+ T-cell cross-priming in a fraction of patients, indicating that montanide contributes to the induction of specific CD8+ T-cell responses to NY-ESO-1.


Subject(s)
Antigens, Neoplasm/administration & dosage , Cancer Vaccines/therapeutic use , Carboxymethylcellulose Sodium/analogs & derivatives , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Mannitol/analogs & derivatives , Melanoma/immunology , Membrane Proteins/administration & dosage , Oleic Acids/administration & dosage , Poly I-C/administration & dosage , Polylysine/analogs & derivatives , Adjuvants, Immunologic/administration & dosage , Aged , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Carboxymethylcellulose Sodium/administration & dosage , Cross-Priming/immunology , Female , Humans , Interferon Inducers/administration & dosage , Male , Mannitol/administration & dosage , Melanoma/therapy , Membrane Proteins/immunology , Middle Aged , Patient Safety , Polylysine/administration & dosage , Skin Neoplasms/immunology , Skin Neoplasms/therapy , Treatment Outcome
8.
Int Rev Cell Mol Biol ; 348: 1-68, 2019.
Article in English | MEDLINE | ID: mdl-31810551

ABSTRACT

Dendritic cells (DCs) are a unique class of immune cells that act as a bridge between innate and adaptive immunity. The discovery of DCs by Cohen and Steinman in 1973 laid the foundation for DC biology, and the advances in the field identified different versions of DCs with unique properties and functions. DCs originate from hematopoietic stem cells, and their differentiation is modulated by Flt3L. They are professional antigen-presenting cells that patrol the environmental interphase, sites of infection, or infiltrate pathological tissues looking for antigens that can be used to activate effector cells. DCs are critical for the initiation of the cellular and humoral immune response and protection from infectious diseases or tumors. DCs can take up antigens using specialized surface receptors such as endocytosis receptors, phagocytosis receptors, and C type lectin receptors. Moreover, DCs are equipped with an array of extracellular and intracellular pattern recognition receptors for sensing different danger signals. Upon sensing the danger signals, DCs get activated, upregulate costimulatory molecules, produce various cytokines and chemokines, take up antigen and process it and migrate to lymph nodes where they present antigens to both CD8 and CD4 T cells. DCs are classified into different subsets based on an integrated approach considering their surface phenotype, expression of unique and conserved molecules, ontogeny, and functions. They can be broadly classified as conventional DCs consisting of two subsets (DC1 and DC2), plasmacytoid DCs, inflammatory DCs, and Langerhans cells.


Subject(s)
Dendritic Cells/classification , Dendritic Cells/cytology , Animals , Dendritic Cells/immunology , Humans
9.
Phys Rev Lett ; 123(8): 081301, 2019 Aug 23.
Article in English | MEDLINE | ID: mdl-31491224

ABSTRACT

We investigate the impact of prior models on the upper bound of the sum of neutrino masses, ∑m_{ν}. Using data from the large scale structure of galaxies, cosmic microwave background, type Ia supernovae, and big bang nucleosynthesis, we argue that cosmological neutrino mass and hierarchy determination should be pursued using exact models, since approximations might lead to incorrect and nonphysical bounds. We compare constraints from physically motivated neutrino mass models (i.e., ones respecting oscillation experiments) to those from models using standard cosmological approximations. The former give a consistent upper bound of ∑m_{ν}≲0.26 eV (95% CI) and yield the first approximation-independent upper bound for the lightest neutrino mass species, m_{0}^{ν}<0.086 eV (95% CI). By contrast, one of the approximations, which is inconsistent with the known lower bounds from oscillation experiments, yields an upper bound of ∑m_{ν}≲0.15 eV (95% CI); this differs substantially from the physically motivated upper bound.

11.
Cell Rep ; 24(7): 1902-1915.e6, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30110645

ABSTRACT

The ability to generate large numbers of distinct types of human dendritic cells (DCs) in vitro is critical for accelerating our understanding of DC biology and harnessing them clinically. We developed a DC differentiation method from human CD34+ precursors leading to high yields of plasmacytoid DCs (pDCs) and both types of conventional DCs (cDC1s and cDC2s). The identity of the cells generated in vitro and their strong homology to their blood counterparts were demonstrated by phenotypic, functional, and single-cell RNA-sequencing analyses. This culture system revealed a critical role of Notch signaling and GM-CSF for promoting cDC1 generation. Moreover, we discovered a pre-terminal differentiation state for each DC type, characterized by high expression of cell-cycle genes and lack of XCR1 in the case of cDC1. Our culture system will greatly facilitate the simultaneous and comprehensive study of primary, otherwise rare human DC types, including their mutual interactions.


Subject(s)
Cell Lineage/immunology , Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Receptor, Notch1/genetics , Antigens, CD34/genetics , Antigens, CD34/immunology , Calcium-Binding Proteins , Cell Cycle Proteins/genetics , Cell Cycle Proteins/immunology , Cell Differentiation/drug effects , Dendritic Cells/cytology , Dendritic Cells/drug effects , Gene Expression , Gene Expression Profiling , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Imidazoles/pharmacology , Immunophenotyping , Intercellular Signaling Peptides and Proteins/immunology , Lipopolysaccharides/pharmacology , Membrane Proteins/immunology , Poly I-C/pharmacology , Primary Cell Culture , Receptor, Notch1/immunology , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , Signal Transduction , Single-Cell Analysis
12.
Cancer J ; 23(2): 131-137, 2017.
Article in English | MEDLINE | ID: mdl-28410301

ABSTRACT

Dendritic cells (DCs) are equipped for sensing danger signals and capturing, processing, and presenting antigens to naive or effector cells and are critical in inducing humoral and adaptive immunity. Successful vaccinations are those that activate DCs to elicit both cellular and humoral responses, as well as long-lasting memory response against the target of interest. Recently, it has become apparent that tumor cells can provide new sources of antigens through nonsynonymous mutations or frame-shift mutations, leading to potentially hundreds of mutation-derived tumor antigens (MTAs) or neoantigens. T cells recognizing MTA have been detected in cancer patients and can even lead to tumor regression. Designing MTA-specific vaccination strategies will have to take into account the adjuvant activity of DC subsets and the best formulation to elicit an effective immune response. We discuss the potential of human DCs to prime MTA-specific responses.


Subject(s)
Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Dendritic Cells/immunology , Mutation , Animals , Humans
13.
J Immunol ; 198(7): 2785-2795, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28228559

ABSTRACT

Targeting Ags to conventional dendritic cells can enhance Ag-specific immune responses. Although most studies have focused on the induction of T cell responses, the mechanisms by which targeting improves Ab responses are poorly understood. In this study we present data on the use of human XCL1 (hXCL1) and hXCL2 fusion vaccines in a murine model. We show that the human chemokines bound type 1 conventional dendritic cells (cDC1), and that immunization with influenza virus hemagglutinin fused to hXCL1 or hXCL2 induced full protection against influenza challenge. Surprisingly, the hXCL1- and hXCL2-fusion vaccines induced better long-term protection associated with stronger induction of neutralizing Abs, and more Ab-secreting cells in bone marrow. In contrast, murine Xcl1 fusion vaccines induced stronger CD8+ T cell responses compared with hXCL1. Further analysis revealed that although murine Xcl1 fusion vaccines induced chemotaxis and were rapidly endocytosed by cDC1, hXCL1 and hXCL2 fusion vaccines did not induce chemotaxis, were less efficiently endocytosed, and consequently, remained on the surface. This difference may explain the enhanced induction of Abs when targeting Ag to cDC1 using hXCL1 and hXCL2, and suggests that immune responses can be manipulated in directing Abs or T cells based on how efficiently the targeted Ag is endocytosed by the DC.


Subject(s)
Dendritic Cells/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza Vaccines/immunology , Receptors, G-Protein-Coupled/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Endocytosis/immunology , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Flow Cytometry , Humans , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology
14.
Cell Res ; 27(1): 74-95, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28025976

ABSTRACT

Immunotherapy using dendritic cell (DC)-based vaccination is an approved approach for harnessing the potential of a patient's own immune system to eliminate tumor cells in metastatic hormone-refractory cancer. Overall, although many DC vaccines have been tested in the clinic and proven to be immunogenic, and in some cases associated with clinical outcome, there remains no consensus on how to manufacture DC vaccines. In this review we will discuss what has been learned thus far about human DC biology from clinical studies, and how current approaches to apply DC vaccines in the clinic could be improved to enhance anti-tumor immunity.


Subject(s)
Dendritic Cells/immunology , Immunotherapy , Animals , Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Dendritic Cells/cytology , Humans , Immunity , Vaccination
15.
Methods Mol Biol ; 1423: 19-37, 2016.
Article in English | MEDLINE | ID: mdl-27142006

ABSTRACT

Dendritic cells (DCs) are a heterogeneous population of professional antigen-presenting cells which play a key role in orchestrating immune defenses. Most of the information gained on human DC biology was derived from studies conducted with DCs generated in vitro from peripheral blood CD14(+) monocytes (MoDCs) or from CD34(+) hematopoietic progenitors. Recent advances in the field revealed that these types of in vitro-derived DCs strikingly differ from the DC subsets that are naturally present in human lymphoid organs, in terms of global gene expression, of specialization in the sensing of different types of danger signals, and of the ability to polarize T lymphocytes toward different functions. Major efforts are being made to better characterize the biology and the functions of lymphoid organ-resident DC subsets in humans, as an essential step for designing innovative DC-based vaccines against infections or cancers. However, this line of research is hampered by the low frequency of certain DC subsets in most tissues, their fragility, and the complexity of the procedures necessary for their purification. Hence, there is a need for robust procedures allowing large-scale in vitro generation of human DC subsets, under conditions allowing their genetic or pharmacological manipulation, to decipher their functions and their molecular regulation. Human CD141(+)CLEC9A(+)XCR1(+) DCs constitute a very interesting DC subset for the design of immunotherapeutic treatments against infections by intracellular pathogens or against cancer, because these cells resemble mouse professional cross-presenting CD8α(+)Clec9a(+)Xcr1(+) DCs. Human XCR1(+) DCs have indeed been reported by several teams to be more efficient than other human DC subsets for cross-presentation, in particular of cell-associated antigens but also of soluble antigens especially when delivered into late endosomes or lysosomes. However, human XCR1(+) DCs are the rarest and perhaps the most fragile of the human DC subsets and hence the most difficult to study ex vivo. Here, we describe a protocol allowing simultaneous in vitro generation of human MoDCs and XCR1(+) DCs, which will undoubtedly be extremely useful to better characterize the functional specialization of human XCR1(+) DCs and to identify its molecular bases.


Subject(s)
Antigens, CD34/metabolism , Dendritic Cells/cytology , Hematopoietic Stem Cells/cytology , Receptors, G-Protein-Coupled/metabolism , Antigens, Surface/metabolism , Cell Differentiation , Cells, Cultured , Cross-Priming , Dendritic Cells/immunology , Flow Cytometry , Hematopoietic Stem Cells/immunology , Humans , In Vitro Techniques , Lectins, C-Type/metabolism , Monocytes/cytology , Monocytes/immunology , Receptors, Mitogen/metabolism , Thrombomodulin
16.
J Immunol ; 193(4): 1622-35, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25009205

ABSTRACT

Human monocyte-derived dendritic cell (MoDC) have been used in the clinic with moderately encouraging results. Mouse XCR1(+) DC excel at cross-presentation, can be targeted in vivo to induce protective immunity, and share characteristics with XCR1(+) human DC. Assessment of the immunoactivation potential of XCR1(+) human DC is hindered by their paucity in vivo and by their lack of a well-defined in vitro counterpart. We report in this study a protocol generating both XCR1(+) and XCR1(-) human DC in CD34(+) progenitor cultures (CD34-DC). Gene expression profiling, phenotypic characterization, and functional studies demonstrated that XCR1(-) CD34-DC are similar to canonical MoDC, whereas XCR1(+) CD34-DC resemble XCR1(+) blood DC (bDC). XCR1(+) DC were strongly activated by polyinosinic-polycytidylic acid but not LPS, and conversely for MoDC. XCR1(+) DC and MoDC expressed strikingly different patterns of molecules involved in inflammation and in cross-talk with NK or T cells. XCR1(+) CD34-DC but not MoDC efficiently cross-presented a cell-associated Ag upon stimulation by polyinosinic-polycytidylic acid or R848, likewise to what was reported for XCR1(+) bDC. Hence, it is feasible to generate high numbers of bona fide XCR1(+) human DC in vitro as a model to decipher the functions of XCR1(+) bDC and as a potential source of XCR1(+) DC for clinical use.


Subject(s)
Antigens, CD34/immunology , Blood Cells/immunology , Dendritic Cells/immunology , Monocytes/immunology , Receptors, G-Protein-Coupled/immunology , Adjuvants, Immunologic/pharmacology , Antigen Presentation/immunology , Cell Culture Techniques , Cell Differentiation/immunology , Cell Line , Cross-Priming/immunology , Gene Expression Profiling , Green Fluorescent Proteins , Humans , Imidazoles/immunology , Killer Cells, Natural/immunology , Lipopolysaccharides/immunology , Phenotype , Poly I-C/immunology , T-Lymphocytes/immunology , Toll-Like Receptor 3 , Toll-Like Receptor 4
17.
Transfusion ; 50(11): 2413-23, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20497510

ABSTRACT

BACKGROUND: Advances in the past two decades in dendritic cell (DC) biology paved the way to exploit them as a promising tool in cancer immunotherapy. The prerequisite for DC vaccine preparations is large-scale in vitro generations of homogeneous, mature, and functional DCs. Frequent improvements are being made in the existing in vitro DC production protocols to achieve this goal. In our previous study we reported a large-scale generation of mature, functional DCs from umbilical cord blood (UCB) CD34+ cells. Here we report that this method can be used for the efficient generation of DCs from UCB mononuclear cells (MNCs) and thus the hematopoietic stem cell isolation step is not essential. STUDY DESIGN AND METHODS: MNCs or CD34+ cells isolated from the same cord blood (CB) samples were used for the generation of DCs. DCs were characterized for morphology, phenotype, and functional assays including antigen uptake, chemotaxis, and mixed leukocyte reaction. Similarly DCs generated from the MNCs of same fresh and frozen CB units were compared. RESULTS: The morphologic, phenotypic, and functional characterization of the DCs generated from various sets show that they were comparable in nature irrespective of the starting population used. CONCLUSION: We conclude that the CD34+ isolation step is not essential for the generation of mature, functional DCs and thus can be eliminated. More importantly, we show that DCs can be generated with equal efficiency from the MNCs of frozen CB units. Our culture method will be useful for exploiting the potential of UCB as an additional source for allogeneic DCs in the clinical settings.


Subject(s)
Blood Preservation , Cell Culture Techniques/methods , Cryopreservation , Dendritic Cells/cytology , Fetal Blood/cytology , Leukocytes, Mononuclear/cytology , Antigens/metabolism , Antigens, CD34/metabolism , Cancer Vaccines , Cell Differentiation/immunology , Chemotaxis, Leukocyte/immunology , Humans , Immunophenotyping , Leukocytes, Mononuclear/metabolism , Lymphocyte Culture Test, Mixed , Receptors, CCR7/metabolism
18.
Transfusion ; 49(10): 2109-21, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19497054

ABSTRACT

BACKGROUND: In vitro generated dendritic cells (DCs) are widely used as adjuvants in cancer immunotherapy. The major sources for DC generation are monocytes and CD34+ cells. CD34+-derived DCs are less frequently used in clinical applications because it requires complex generation methods. Here a simple method for the large-scale generation of mature functional DCs from umbilical cord blood-derived CD34+ cells is described. STUDY DESIGN AND METHODS: CD34+ cells were first expanded with a combination of early acting growth factors in a medium containing autologous plasma. In the second step the DC precursors were further either enriched by plastic adherence or sorted on a cell sorter and differentiated as DCs. DCs generated by both methods were compared for their morphology, phenotype, and different functional variables. RESULTS: This culture system provided a large-scale expansion of CD34+ cells giving a mean fold increase of 615. The majority of the expanded cells were interstitial DC precursors, that is, CD14+-positive cells. In vitro generated immature DCs could be matured into functional DCs by appropriate maturation stimuli. DCs generated by the plastic adherence method had a better cytokine profile and strong mixed leukocyte reaction compared to those generated by cell sorting. CONCLUSION: A two-step culture system provides a large-scale expansion of CD34+ cells with a preferential lineage commitment toward CD14+ cells. Enrichment of these precursors with a simple plastic adherence technique results in generation of large numbers of mature, functional DCs. This method of in vitro DC generation will have applications in cancer immunotherapy.


Subject(s)
Antigens, CD34/immunology , Cell Culture Techniques/methods , Cell Differentiation , Dendritic Cells/cytology , Dendritic Cells/immunology , Fetal Blood/cytology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Interleukin-10/metabolism , Interleukin-12/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...