Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 16(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38893237

ABSTRACT

In this review, we aim to provide a summary of the diverse immunophenotypic presentations of distinct entities associated with plasmacytoid dendritic cell (pDC) proliferation. These entities include the following: (1) blastic plasmacytoid dendritic cell neoplasm (BPDCN); (2) mature pDC proliferation (MPDCP), most commonly seen in chronic myelomonocytic leukemia (CMML); and (3) myeloid neoplasms with pDC differentiation, in which pDCs show a spectrum of maturation from early immature pDCs to mature forms, most commonly seen in acute myeloid leukemia (pDC-AML). Our aim is to provide a flow cytometry diagnostic approach to these distinct and sometimes challenging entities and to clarify the immunophenotypic spectrum of neoplastic pDCs in different disease presentations. In this review, we also cover the strategies in the evaluation of residual disease, as well as the challenges and pitfalls we face in the setting of immune and targeted therapy. The differential diagnosis will also be discussed, as blasts in some AML cases can have a pDC-like immunophenotype, mimicking pDCs.

2.
Trop Med Infect Dis ; 8(10)2023 Sep 26.
Article in English | MEDLINE | ID: mdl-37888588

ABSTRACT

In an attempt to find new anti-echinococcal drugs, resveratrol (Rsv) effectiveness against the larval stages of Echinococcus granulosus and E. multilocularis was evaluated. The in vitro effect of Rsv on parasites was assessed via optical and electron microscopy, RT-qPCR and immunohistochemistry. In vivo efficacy was evaluated in murine models of cystic (CE) and alveolar echinococcosis (AE). The impact of infection and drug treatment on the mouse bone marrow hematopoietic stem cell (HSC) population and its differentiation into dendritic cells (BMDCs) was investigated via flow cytometry and RT-qPCR. In vitro treatment with Rsv reduced E. granulosus metacestode and protoscolex viability in a concentration-dependent manner, caused ultrastructural damage, increased autophagy gene transcription, and raised Eg-Atg8 expression while suppressing Eg-TOR. However, the intraperitoneal administration of Rsv was not only ineffective, but also promoted parasite development in mice with CE and AE. In the early infection model of AE treated with Rsv, an expansion of HSCs was observed followed by their differentiation towards BMCDs. The latter showed an anti-inflammatory phenotype and reduced LPS-stimulated activation compared to control BMDCs. We suggest that Rsv ineffectiveness could have been caused by the low intracystic concentration achieved in vivo and the drug's hormetic effect, with opposite anti-parasitic and immunomodulatory responses in different doses.

3.
Cell Immunol ; 349: 104043, 2020 03.
Article in English | MEDLINE | ID: mdl-32044112

ABSTRACT

Type I Interferon (IFN) signaling plays a critical role in dendritic cell (DC) development and functions. Inhibition of hyper type I IFN signaling promotes cDC2 subtype development. Relb is essential to development of cDC2 subtype and here we analyzed its effect on type I IFN signaling in DCs. We show that Relb suppresses the homeostatic type I IFN signaling in cDC2 cultures. TLR stimulation of FL-DCs led to RelB induction coinciding with fall in IFN signatures; conforming with the observation Relb expression reduced TLR stimulated IFN induction along with decrease in ISGs. Towards understanding mechanism, we show that effects of RelB are mediated by increased levels of IκBα. We demonstrate that RelB dampened antiviral responses by lowering ISG levels and the defect in cDC2 development in RelB null mice can be rescued in Ifnar1-/- background. Overall, we propose a novel role of RelB as a negative regulator of the type I IFN signaling pathway; fine tuning development of cDC2 subtype.


Subject(s)
Dendritic Cells/immunology , Interferon Type I/immunology , NF-KappaB Inhibitor alpha/physiology , Transcription Factor RelB/physiology , Amino Acid Sequence , Animals , Cell Differentiation , Cells, Cultured , Crosses, Genetic , Dendritic Cells/classification , Dendritic Cells/cytology , Gene Expression Regulation/immunology , Mice , NIH 3T3 Cells , Newcastle disease virus/immunology , Peptides/pharmacology , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/physiology , Signal Transduction/immunology , Spleen/cytology , Transcription Factor RelB/deficiency , Transcription Factor RelB/genetics , Viral Load
4.
Front Immunol ; 11: 593363, 2020.
Article in English | MEDLINE | ID: mdl-33613517

ABSTRACT

Dendritic cell (DC)-based antitumor vaccines have proven to be a safe approach, but often fail to generate robust results between trials. Translation to the clinic has been hindered in part by the lack of standard operation procedures for vaccines production, namely the definition of optimal culture conditions during ex-vivo DC differentiation. Here we sought to compare the ability of three clinical grade serum-free media, DendriMACS, AIM-V, and X-VIVO 15, alongside with fetal bovine serum-supplemented Roswell Park Memorial Institute Medium (RPMI), to support the differentiation of monocyte-derived DCs (Mo-DCs). Under these different culture conditions, phenotype, cell metabolomic profiles, response to maturation stimuli, cytokines production, allogenic T cell stimulatory capacity, as well as priming of antigen-specific CD8+ T cells and activation of autologous natural killer (NK) cells were analyzed. Immature Mo-DCs differentiated in AIM-V or X-VIVO 15 presented lower levels of CD1c, CD1a, and higher expression of CD11c, when compared to cells obtained with DendriMACS. Upon stimulation, only AIM-V or X-VIVO 15 DCs acquired a full mature phenotype, which supports their enhanced capacity to polarize T helper cell type 1 subset, to prime antigen-specific CD8+ T cells and to activate NK cells. CD8+ T cells and NK cells resulting from co-culture with AIM-V or X-VIVO 15 DCs also showed superior cytolytic activity. 1H nuclear magnetic resonance-based metabolomic analysis revealed that superior DC immunostimulatory capacities correlate with an enhanced catabolism of amino acids and glucose. Overall, our data highlight the impact of critically defining the culture medium used in the production of DCs for clinical application in cancer immunotherapy. Moreover, the manipulation of metabolic state during differentiation could be envisaged as a strategy to enhance desired cell characteristics.


Subject(s)
Batch Cell Culture Techniques , Culture Media, Serum-Free , Dendritic Cells/immunology , Dendritic Cells/metabolism , Immunotherapy , Primary Cell Culture/methods , Batch Cell Culture Techniques/methods , Batch Cell Culture Techniques/standards , Biomarkers , Cell Differentiation , Cytokines/metabolism , Cytotoxicity Tests, Immunologic , Dendritic Cells/cytology , Humans , Immunophenotyping , Immunotherapy/methods , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocyte Activation/immunology , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Phagocytosis , Primary Cell Culture/standards
5.
Front Microbiol ; 10: 2445, 2019.
Article in English | MEDLINE | ID: mdl-31736892

ABSTRACT

The cell wall has a critical role in the host immune response to fungal pathogens. In this study, we investigated the influence of two cell wall fractions of the dimorphic fungi Paracoccidioides brasiliensis (Pb) in the in vitro generation of monocyte-derived dendritic cells (MoDCs). Monocytes were purified from the peripheral blood of healthy donors and cultivated for 7 days in medium supplemented with IL-4 and GM-CSF in the presence of Pb cell wall fractions: the alkali-insoluble F1, constituted by ß-1,3-glucans, chitin and proteins, and the alkali-soluble F2, mainly constituted by α-glucan. MoDCs phenotypes were evaluated regarding cell surface expression of CD1a, DC-SIGN, HLA-DR, CD80, and CD83 and production of cytokines. The α-glucan-rich cell wall fraction downregulated the differentiation of CD1a+ MoDCs, a dendritic cell subset that stimulate Th1 responses. The presence of both cell fractions inhibited DC-SIGN and HLA-DR expression, while the expression of maturation markers was differentially induced in CD1a- MoDCs. Differentiation upon F1 and F2 stimulation induced mixed profile of inflammatory cytokines. Altogether, these data demonstrate that Pb cell wall fractions differentially induce a dysregulation in DCs differentiation. Moreover, our results suggest that cell wall α-glucan promote the differentiation of CD1a- DCs, potentially favoring Th2 polarization and contributing to pathogen persistence.

6.
J Clin Med ; 7(10)2018 Sep 29.
Article in English | MEDLINE | ID: mdl-30274241

ABSTRACT

Clinical application of tissue engineering products requires the exclusion of immune responses after implantation. We used jaw periosteal cells (JPCs) as a suitable stem cell source and analyzed herein the effects of JPCs on dendritic cell maturation after co-culturing of both cell types. Peripheral blood mononuclear cells (PBMCs) were differentiated to dendritic cells (DCs) by the addition of differentiation cocktails for 7 days in co-culture with undifferentiated and osteogenically induced JPCs. The effects of JPCs on DC maturation were analyzed at the beginning (day 7), in the middle (day 14), and at the end (day 21) of the osteogenesis process. We detected significantly lower DC numbers after co-culturing with JPCs that have previously been left untreated or osteogenically differentiated for 7, 14, and 21 days. Using gene expression analyses, significantly lower IL-12p35 and -p40 and pro-inflammatory cytokine (IFN-γ and TNF-α) levels were detected, whereas IL-8 mRNA levels were significantly higher in DCs. Furthermore, osteogenic media conditions enhanced significantly IL-10 gene expression. We concluded that undifferentiated and osteogenically differentiated JPCs had an overall inhibiting influence on dendritic cell maturation. Further studies should clarify the underlaying mechanism in depth.

7.
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
8.
Bio Protoc ; 7(15): e2419, 2017 Aug 05.
Article in English | MEDLINE | ID: mdl-34541147

ABSTRACT

Induced pluripotent stem cells (iPS cells) are engineered stem cells, which exhibit properties very similar to embryonic stem cells (ES cells; Takahashi and Yamanaka, 2016). Both iPS cells and ES cells have an extraordinary self-renewal capacity and can differentiate into all cell types of our body, including hematopoietic stem/progenitor cells and dendritic cells (DC) derived thereof. This makes iPS cells particularly well suited for studying molecular mechanisms of diseases, drug discovery and regenerative therapy ( Grskovic et al., 2011 ; Bellin et al., 2012 ; Robinton and Daley, 2012). DC are the major antigen presenting cells of the immune system and thus they are key players in modulating and directing immune responses ( Merad et al., 2013 ). DC patrol peripheral and interface tissues (e.g., lung, intestine and skin) to detect invading pathogens, and upon activation they migrate to lymph nodes to activate and prime lymphocytes. DC comprise a phenotypically heterogeneous family with functionally specialized subsets (Schlitzer and Ginhoux, 2014). Generally, classical DC (cDC) and plasmacytoid DC (pDC) are distinguished, exhibiting a classical and plasma cell-like DC morphology, respectively. cDC recognize a multitude of pathogens and secrete proinflammatory cytokines upon activation, while pDC are specialized to detect intracellular pathogens and secrete type I interferons ( Merad et al., 2013 ; Schlitzer and Ginhoux, 2014). cDC are further divided into cross-presenting cDC1 and conventional cDC2, in the human system referred to as CD141+ Clec9a+ cDC1 and CD1c+ CD14- cDC2. Human pDC are characterized as CD303+ CD304+ ( Jongbloed et al., 2010 ; Joffre et al., 2012 ; Swiecki and Colonna, 2015). To investigate subset specification and function of human DC, we established a protocol to generate cDC1, cDC2 and pDC in vitro from human iPS cells (or ES cells) ( Sontag et al., 2017 ). Therefore, we differentiated iPS cells (or ES cells), via mesoderm commitment and hemato-endothelial specification, into CD43+ CD31+ hematopoietic progenitors. Subsequently, those were seeded onto inactivated OP9 stromal cells with FLT3L, SCF, GM-CSF and IL-4 or FLT3L, SCF and GM-CSF to specify cDC1 and cDC2, or cDC1 and pDC, respectively.

9.
Oncotarget ; 7(21): 30193-210, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27070086

ABSTRACT

Human mesenchymal stromal cells (MSC) have been shown to dampen immune response and promote tissue repair, but the underlying mechanisms are still under investigation. Herein, we demonstrate that umbilical cord-derived MSC (UC-MSC) alter the phenotype and function of monocyte-derived dendritic cells (DC) through lactate-mediated metabolic reprogramming. UC-MSC can secrete large quantities of lactate and, when present during monocyte-to-DC differentiation, induce instead the acquisition of M2-macrophage features in terms of morphology, surface markers, migratory properties and antigen presentation capacity. Microarray expression profiling indicates that UC-MSC modify the expression of metabolic-related genes and induce a M2-macrophage expression signature. Importantly, monocyte-derived DC obtained in presence of UC-MSC, polarize naïve allogeneic CD4+ T-cells into Th2 cells. Treatment of UC-MSC with an inhibitor of lactate dehydrogenase strongly decreases lactate concentration in culture supernatant and abrogates the effect on monocyte-to-DC differentiation. Metabolic analysis further revealed that UC-MSC decrease oxidative phosphorylation in differentiating monocytes while strongly increasing the spare respiratory capacity proportional to the amount of secreted lactate. Because both MSC and monocytes are recruited in vivo at the site of tissue damage and inflammation, we propose the local increase of lactate concentration induced by UC-MSC and the consequent enrichment in M2-macrophage generation as a mechanism to achieve immunomodulation.


Subject(s)
Cell Differentiation/genetics , Lactic Acid/metabolism , Macrophages/metabolism , Mesenchymal Stem Cells/metabolism , Animals , Cell Differentiation/drug effects , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/metabolism , Gene Expression Profiling/methods , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Interleukin-4/pharmacology , Macrophages/cytology , Mice, Inbred C57BL , Mice, SCID , Monocytes/cytology , Monocytes/metabolism , Umbilical Cord/cytology
10.
Microbiol Immunol ; 59(12): 724-34, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26564804

ABSTRACT

Mannan-binding lectin (MBL), a circulating C-type lectin, is an important member of the defense collagen family. It exhibits a high potential for recognizing broad categories of pathogen-associated molecular patterns and initiating complement cascade responses. DCs are well-known specialist antigen-presenting cells that significantly trigger specific T cell-mediated immune responses. In our previous study, it was observed that high concentrations of MBL significantly attenuate LPS-induced maturation of monocyte-derived DCs (MoDCs). In the current study, it was postulated that MBL at similar supraphysiological concentrations would affect early differentiation of MoDCs in some way. CD14(+) monocytes from human peripheral blood mononuclear cells were cultured with granulocyte-macrophage colony-stimulating factor and IL-4 in the presence or absence of physiological (1 µg/mL) and supraphysiological concentrations (20 µg/mL) of MBL protein, respectively. Phenotypic analysis indicated that the differentiated DCs incubated with high concentrations of MBL expressed MHC class II and costimulatory molecules (e.g., CD80 and CD40) more weakly than did control groups. The secretion of IL-10 and IL-6 increased markedly, whereas their mixed lymphocyte reaction-stimulating capacity decreased. Members of the signal transducer and activator of transcription family were also found to be differentially regulated. Thus, beyond the role of MBL as an opsonin, our data reveal a possible inhibitory effect of MBL at high concentrations in monocyte-DC transition, which probably provides one way of regulating adaptive immune responses by strict regulation of DCs, making MBL a better prospect for controlling relevant pathological events such as autoimmune diseases.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/drug effects , Leukocytes, Mononuclear/drug effects , Lipopolysaccharide Receptors/biosynthesis , Mannose-Binding Lectin/pharmacology , Monocytes/drug effects , Apoptosis/drug effects , Blotting, Western , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Interleukin-10/biosynthesis , Interleukin-4/pharmacology , Interleukin-6/biosynthesis , Interleukin-6/metabolism , Leukocytes, Mononuclear/cytology , Lipopolysaccharide Receptors/immunology , Monocytes/cytology , Monocytes/immunology , Phenotype , Protein Binding , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
11.
J Leukoc Biol ; 96(6): 1001-10, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25170118

ABSTRACT

This study tests the hypothesis that CD8α(+) DCs in the spleen of mice contain an immature precursor for functionally mature, "classical" cross-presenting CD8α(+) DCs. The lymphoid tissues contain a network of phenotypically distinct DCs with unique roles in surveillance and immunity. Splenic CD8α(+) DCs have been shown to exhibit a heightened capacity for phagocytosis of cellular material, secretion of IL-12, and cross-priming of CD8(+) T cells. However, this population can be subdivided further on the basis of expression of both langerin/CD207 and CX(3)CR1. We therefore evaluated the functional capacities of these different subsets. The CX(3)CR1(+) CD8α(+) DC subset does not express langerin and does not exhibit the classical features above. The CX(3)CR1(-) CD8α(+) DC can be divided into langerin-positive and negative populations, both of which express DEC205, Clec9A, and high basal levels of CD86. However, the langerin(+) CX(3)CR1(-) CD8α(+) subset has a superior capacity for acquiring cellular material and producing IL-12 and is more susceptible to activation-induced cell death. Significantly, following purification and adoptive transfer into new hosts, the langerin(-) CX(3)CR1(-) CD8α(+) subset survives longer, up-regulates expression of langerin, and becomes more susceptible to activation-induced cell death. Last, in contrast to langerin(+) CX(3)CR1(-) CD8α(+), the langerin(-) CX(3)CR1(-) CD8α(+) are still present in Batf3(-/-) mice. We conclude that the classical attributes of CD8α(+) DC are confined primarily to the langerin(+) CX(3)CR1(-) CD8α(+) DC population and that the langerin(-) CX(3)CR1(-) subset represents a Batf3-independent precursor to this mature population.


Subject(s)
Adaptive Immunity , Antigens, Differentiation/analysis , Dendritic Cells/classification , Adoptive Transfer , Animals , Antigen Presentation , Antigens, Surface/analysis , Antigens, Surface/biosynthesis , Antigens, Surface/genetics , Basic-Leucine Zipper Transcription Factors/analysis , CD8 Antigens/analysis , CD8-Positive T-Lymphocytes/immunology , CX3C Chemokine Receptor 1 , Cell Differentiation , Cell Lineage , Cells, Cultured , Cellular Senescence , Crosses, Genetic , Dendritic Cells/chemistry , Dendritic Cells/immunology , Female , Galactosylceramides/immunology , Histocompatibility Antigens Class I/immunology , Immune Tolerance/immunology , Immunophenotyping , Interleukin-12 Subunit p40/biosynthesis , Lectins, C-Type/analysis , Lectins, C-Type/biosynthesis , Lectins, C-Type/genetics , Male , Mannose-Binding Lectins/analysis , Mannose-Binding Lectins/biosynthesis , Mannose-Binding Lectins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phagocytosis/immunology , Receptors, Chemokine/analysis , Repressor Proteins/analysis , Spleen/cytology , Spleen/immunology
12.
Hum Immunol ; 75(7): 641-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24768898

ABSTRACT

Professional antigen-presenting cells, dendritic cells (DCs) play an important role in controlling tumors. It is known that solid tumor cell products inhibit DC differentiation. Recently a similar effect produced by leukemic cell products has been demonstrated. In this case, leukemic cell products induced the secretion of IL-1ß by monocytes undergoing differentiation. The aim of the present work was to characterize and to compare the development of monocyte-derived DCs under the influence of leukemic cell products (K562 supernatant) or exogenous IL-1ß. It became clear that leukemic cell products and IL-1ß differentially modulate some of the parameters studied on monocytes stimulated to differentiate into DCs. In the presence of K562 supernatant, the expression of the macrophage markers CD16 and CD68 were higher than in immature DCs control. Contrasting with IL-1ß, leukemic cell products possibly favor the development of cells with macrophage markers. In addition, CD80 and CD83 expressions were also higher in the presence of tumor supernatant whereas HLA-DR was lower. In the presence of IL-1ß, only CD80 was increased. Furthermore, it was observed that when monocytes were induced to differentiate into DCs in the presence of tumor supernatant and then activated, they expressed less CD80 and CD83 than activated DCs control. A reduced expression of CD83 following activation was also seen in cells differentiated with IL-1ß. TGF-ß and VEGF were found in the tumor supernatants. Moreover, the exposure to tumor supernatant or IL-1ß stimulated IL-10 production while decreased IL-12 production by activated DCs. Finally, these results suggest that the addition of products released by leukemic cells or, more discreetly, the addition of IL-1ß affects DC differentiation, inducing a suppressive phenotype.


Subject(s)
Culture Media, Conditioned/pharmacology , Dendritic Cells/drug effects , Interleukin-1beta/pharmacology , Monocytes/drug effects , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/immunology , B7-1 Antigen/genetics , B7-1 Antigen/immunology , Cell Communication , Cell Differentiation , Coculture Techniques , Dendritic Cells/cytology , Dendritic Cells/immunology , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Gene Expression , Genotype , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , HLA-DR Antigens/genetics , HLA-DR Antigens/immunology , Humans , Immunoglobulins/genetics , Immunoglobulins/immunology , Interleukin-10/biosynthesis , Interleukin-10/metabolism , Interleukin-12/biosynthesis , Interleukin-12/metabolism , Interleukin-4/pharmacology , K562 Cells , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Monocytes/cytology , Monocytes/immunology , Phenotype , Primary Cell Culture , Receptors, IgG/genetics , Receptors, IgG/immunology , Tumor Necrosis Factor-alpha/pharmacology , CD83 Antigen
13.
Oncoimmunology ; 2(12): e26744, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24575378

ABSTRACT

We observed a cell concentration-dependent differentiation switch among cultured dendritic cells (DCs) triggered by lactic acid, a product of glycolytic metabolism. In particular, while interleukin (IL)-12, IL-23, and tumor necrosis factor α (TNFα)-producing, migratory DCs developed in sparse cultures, IL-10-producing, non-migratory DCs differentiated in dense cultures. This points to a novel opportunity for tailoring DC-based anticancer therapies through metabolism modulation in developing DCs.

SELECTION OF CITATIONS
SEARCH DETAIL