Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Front Immunol ; 8: 1097, 2017.
Article in English | MEDLINE | ID: mdl-28936211

ABSTRACT

Macrophages (Mf) are a heterogeneous population of tissue-resident professional phagocytes and a major component of the leukocyte infiltrate at sites of inflammation, infection, and tumor growth. They can undergo diverse forms of activation in response to environmental factors, polarizing into specialized functional subsets. A common hallmark of the pathologic environment is represented by hypoxia. The impact of hypoxia on human Mf polarization has not been fully established. The objective of this study was to elucidate the effects of a hypoxic environment reflecting that occurring in vivo in diseased tissues on the ability of human Mf to polarize into classically activated (proinflammatory M1) and alternatively activated (anti-inflammatory M2) subsets. We present data showing that hypoxia hinders Mf polarization toward the M1 phenotype by decreasing the expression of T cell costimulatory molecules and chemokine homing receptors and the production of proinflammatory, Th1-priming cytokines typical of classical activation, while promoting their acquisition of phenotypic and secretory features of alternative activation. Furthermore, we identify the triggering receptor expressed on myeloid cells (TREM)-1, a member of the Ig-like immunoregulatory receptor family, as a hypoxia-inducible gene in Mf and demonstrate that its engagement by an agonist Ab reverses the M2-polarizing effect of hypoxia imparting a M1-skewed phenotype to Mf. Finally, we provide evidence that Mf infiltrating the inflamed hypoxic joints of children affected by oligoarticular juvenile idiopatic arthritis express high surface levels of TREM-1 associated with predominant M1 polarization and suggest the potential of this molecule in driving M1 proinflammatory reprogramming in the hypoxic synovial environment.

2.
J Mol Med (Berl) ; 94(8): 943-55, 2016 08.
Article in English | MEDLINE | ID: mdl-26960761

ABSTRACT

UNLABELLED: Langerhans cells (LCs) are a specialized dendritic cell subset that resides in the epidermis and mucosal epithelia and is critical for the orchestration of skin immunity. Recent evidence suggest that LCs are involved in aberrant wound healing and in the development of hypertrophic scars and chronic wounds, which are characterized by a hypoxic environment. Understanding LCs biology under hypoxia may, thus, lead to the identification of novel pathogenetic mechanisms of wound repair disorders and open new therapeutic opportunities to improve wound healing. In this study, we characterize a previously unrecognized role for hypoxia in significantly affecting the phenotype and functional properties of human monocyte-derived LCs, impairing their ability to stimulate naive T cell responses, and identify the triggering receptor expressed on myeloid (TREM)-1, a member of the Ig immunoregulatory receptor family, as a new hypoxia-inducible gene in LCs and an activator of their proinflammatory and Th1-polarizing functions in a hypoxic environment. Furthermore, we provide the first evidence of TREM-1 expression in vivo in LCs infiltrating hypoxic areas of active hypertrophic scars and decubitous ulcers, pointing to a potential pathogenic role of this molecule in wound repair disorders. KEY MESSAGES: Hypoxia modulates surface molecule expression and cytokine profile in Langerhans cells. Hypoxia impairs human Langerhans cell stimulatory activity on naive T cells. Hypoxia selectively induces TREM-1 expression in human Langerhans cells. TREM-1 engagement stimulates Langerhans cell inflammatory and Th1-polarizing activity. TREM-1 is expressed in vivo in Langerhans cells infiltrating hypoxic skin lesions.


Subject(s)
Langerhans Cells/physiology , Cell Hypoxia , Cell Proliferation , Cells, Cultured , Cicatrix, Hypertrophic/immunology , Cicatrix, Hypertrophic/metabolism , Cicatrix, Hypertrophic/pathology , Cytokines/metabolism , Humans , Lymphocyte Activation , Skin/immunology , Skin/pathology , T-Lymphocytes/physiology , Triggering Receptor Expressed on Myeloid Cells-1/metabolism
3.
Clin Cancer Res ; 20(11): 2910-21, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24668647

ABSTRACT

PURPOSE: Despite the great success of HER2 vaccine strategies in animal models, effective clinical results have not yet been obtained. We studied the feasibility of using DNA coding for chimeric rat/human HER2 as a tool to break the unresponsiveness of T cells from patients with HER2-overexpressing tumors (HER2-CP). EXPERIMENTAL DESIGN: Dendritic cells (DCs) generated from patients with HER2-overexpressing breast (n = 28) and pancreatic (n = 16) cancer were transfected with DNA plasmids that express human HER2 or heterologous rat sequences in separate plasmids or as chimeric constructs encoding rat/human HER2 fusion proteins and used to activate autologous T cells. Activation was evaluated by IFN-γ ELISPOT assay, perforin expression, and ability to halt HER2+ tumor growth in vivo. RESULTS: Specific sustained proliferation and IFN-γ production by CD4 and CD8 T cells from HER2-CP was observed after stimulation with autologous DCs transfected with chimeric rat/human HER2 plasmids. Instead, T cells from healthy donors (n = 22) could be easily stimulated with autologous DCs transfected with any human, rat, or chimeric rat/human HER2 plasmid. Chimeric HER2-transfected DCs from HER2-CP were also able to induce a sustained T-cell response that significantly hindered the in vivo growth of HER2(+) tumors. The efficacy of chimeric plasmids in overcoming tumor-induced T-cell dysfunction relies on their ability to circumvent suppressor effects exerted by regulatory T cells (Treg) and/or interleukin (IL)-10 and TGF-ß1. CONCLUSIONS: These results provide the proof of concept that chimeric rat/human HER2 plasmids can be used as effective vaccines for any HER2-CP with the advantage of being not limited to specific MHC. Clin Cancer Res; 20(11); 2910-21. ©2014 AACR.


Subject(s)
Breast Neoplasms/immunology , Cancer Vaccines/immunology , Pancreatic Neoplasms/immunology , Receptor, ErbB-2/immunology , Vaccines, DNA/immunology , Animals , Cancer Vaccines/pharmacology , Dendritic Cells/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Humans , Immune Tolerance/immunology , Immunohistochemistry , Mice , Mice, Inbred NOD , Mice, SCID , Plasmids , Rats , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/pharmacology , Transfection , Transplantation Chimera , Vaccines, DNA/pharmacology , Xenograft Model Antitumor Assays
4.
PLoS One ; 8(3): e59403, 2013.
Article in English | MEDLINE | ID: mdl-23527185

ABSTRACT

UNLABELLED: The objective of the present study was to evaluate whether placental mesenchymal stromal cells (PDMSCs) derived from normal and preeclamptic (PE) chorionic villous tissue presented differences in their cytokines expression profiles. Moreover, we investigated the effects of conditioned media from normal and PE-PDMSCs on the expression of pro-inflammatory Macrophage migration Inhibitory Factor (MIF), Vascular Endothelial Growth Factor (VEGF), soluble FMS-like tyrosine kinase-1 (sFlt-1) and free ß-human Chorionic Gonadotropin (ßhCG) by normal term villous explants. This information will help to understand whether anomalies in PE-PDMSCs could cause or contribute to the anomalies typical of preeclampsia. METHODS: Chorionic villous PDMSCs were isolated from severe preeclamptic (n = 12) and physiological control term (n = 12) placentae. Control and PE-PDMSCs's cytokines expression profiles were determined by Cytokine Array. Control and PE-PDMSCs were plated for 72 h and conditioned media (CM) was collected. Physiological villous explants (n = 48) were treated with control or PE-PDMSCs CM for 72 h and processed for mRNA and protein isolation. MIF, VEGF and sFlt-1 mRNA and protein expression were analyzed by Real Time PCR and Western Blot respectively. Free ßhCG was assessed by immunofluorescent. RESULTS: Cytokine array showed increased release of pro-inflammatory cytokines by PE relative to control PDMSCs. Physiological explants treated with PE-PDMSCs CM showed significantly increased MIF and sFlt-1 expression relative to untreated and control PDMSCs CM explants. Interestingly, both control and PE-PDMSCs media induced VEGF mRNA increase while only normal PDMSCs media promoted VEGF protein accumulation. PE-PDMSCs CM explants released significantly increased amounts of free ßhCG relative to normal PDMSCs CM ones. CONCLUSIONS: Herein, we reported elevated production of pro-inflammatory cytokines by PE-PDMSCs. Importantly, PE PDMSCs induced a PE-like phenotype in physiological villous explants. Our data clearly depict chorionic mesenchymal stromal cells as central players in placental physiopathology, thus opening to new intriguing perspectives for the treatment of human placental-related disorders as preeclampsia.


Subject(s)
Chorionic Villi/metabolism , Cytokines/metabolism , Mesenchymal Stem Cells/metabolism , Placenta/cytology , Pre-Eclampsia/etiology , Pre-Eclampsia/physiopathology , Blotting, Western , Chorionic Gonadotropin/metabolism , Female , Fluorescent Antibody Technique , Humans , Macrophage Migration-Inhibitory Factors/metabolism , Pregnancy , Protein-Tyrosine Kinases/metabolism , Real-Time Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/metabolism
5.
Eur J Immunol ; 43(4): 949-66, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23436478

ABSTRACT

DCs are powerful antigen-presenting cells central in the orchestration of innate and acquired immunity. DC development, migration, and activities are intrinsically linked to the microenvironment. DCs migrate through pathologic tissues before reaching their final destination in the lymph nodes. Hypoxia, a condition of low partial oxygen pressure, is a common feature of many pathologic situations, capable of modifying DC phenotype and functional behavior. We studied human monocyte-derived immature DCs generated under chronic hypoxic conditions (H-iDCs). We demonstrate by gene expression profiling the upregulation of a cluster of genes coding for antigen-presentation, immunoregulatory, and pattern recognition receptors, suggesting a stimulatory role for hypoxia on iDC immunoregulatory functions. In particular, we show that H-iDCs express triggering receptor expressed on myeloid cells(TREM-1), a member of the Ig superfamily of immunoreceptors and an amplifier of inflammation. This effect is reversible because H-iDC reoxygenation results in TREM-1 down-modulation. TREM-1 engagement promotes upregulation of T-cell costimulatory molecules and homing chemokine receptors, typical of mature DCs, and increases the production of proinflammatory, Th1/Th17-priming cytokines/chemokines, resulting in increased T-cell responses. These results suggest that TREM-1 induction by the hypoxic microenvironment represents a mechanism of regulation of Th1-cell trafficking and activation by iDCs differentiated at pathologic sites.


Subject(s)
Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Inflammation Mediators/metabolism , Membrane Glycoproteins/metabolism , Phenotype , Receptors, Immunologic/metabolism , Cell Hypoxia , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation , Humans , Membrane Glycoproteins/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Triggering Receptor Expressed on Myeloid Cells-1
6.
Immunobiology ; 218(1): 76-89, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22465745

ABSTRACT

Myeloid dendritic cells (DCs) are professional antigen-presenting cells critical for the orchestration of immunity and maintenance of self-tolerance. DC development and functions are tightly regulated by a complex network of inhibitory and activating signals present in the tissue microenvironment, and dysregulated DC responses may result in amplification of inflammation, loss of tolerance, or establishment of immune escape mechanisms. Generation of mature (m)DCs from monocytic precursors recruited at pathological sites occurs under condition of low partial oxygen pressure (pO(2)). However, the way in which the hypoxic microenvironment modulates the functions of these cells is still not clear. We demonstrate that chronic hypoxia (4 days, 1% O(2)) promotes the onset of a highly proinflammatory gene expression profile in mDCs generated from primary human monocytes, characterized by the modulation of a significant cluster of genes coding for proinflammatory chemokines/cytokines and/or their receptors. Within the chemokine system, strong upregulation of genes encoding proteins chemotactic for neutrophils, such as CXCL2, CXCL3, CXCL5, CXCL6, and CXCL8, and for activated/memory T lymphocytes, monocytes, and immature (i) DCs, e.g. CCL20, CCL3 and CCL5, was observed, concomitant with decreased expression of genes coding for naive/resting T cells chemoattractants, CCL18 and CCL23. Other hypoxia-inducible genes coded for cytokines with a primary role in inflammation and angiogenesis, including osteopontin, vascular endothelial growth factor, and IL-1ß. mRNA modulation was paralleled by protein secretion. These results suggest that conditions of reduced O(2) availability reprograms mDCs toward a proinflammatory direction by tuning the cytokine/chemokine repertoire, thus affecting their ability to regulate leukocyte trafficking and activation at pathological sites, with potential implications for the pathogenesis of chronic inflammatory diseases.


Subject(s)
Cell Hypoxia/immunology , Chemokines/metabolism , Cytokines/metabolism , Dendritic Cells/immunology , Osteopontin/metabolism , Cell Differentiation , Cell Movement , Cells, Cultured , Cellular Microenvironment/immunology , Chemokines/genetics , Cytokines/genetics , Gene Expression Regulation/immunology , Humans , Immunity, Cellular/genetics , Inflammation Mediators/metabolism , Monocytes/immunology , Neovascularization, Pathologic/genetics , Osteogenesis/genetics , Osteopontin/genetics , Transcriptome , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
7.
Blood ; 117(9): 2625-39, 2011 Mar 03.
Article in English | MEDLINE | ID: mdl-21148811

ABSTRACT

Dendritic cells (DCs) are a heterogeneous group of professional antigen-presenting cells functioning as sentinels of the immune system and playing a key role in the initiation and amplification of innate and adaptive immune responses. DC development and functions are acquired during a complex differentiation and maturation process influenced by several factors present in the local milieu. A common feature at pathologic sites is represented by hypoxia, a condition of low pO(2), which creates a unique microenvironment affecting cell phenotype and behavior. Little is known about the impact of hypoxia on the generation of mature DCs (mDCs). In this study, we identified by gene expression profiling a significant cluster of genes coding for immune-related cell surface receptors strongly up-regulated by hypoxia in monocyte-derived mDCs and characterized one of such receptors, TREM-1, as a new hypoxia-inducible gene in mDCs. TREM-1 associated with DAP12 in hypoxic mDCs, and its engagement elicited DAP12-linked signaling, resulting in ERK-1, Akt, and IκBα phosphorylation and proinflammatory cytokine and chemokine secretion. Finally, we provided the first evidence that TREM-1 is expressed on mDCs infiltrating the inflamed hypoxic joints of children affected by juvenile idiopathic arthritis, representing a new in vivo marker of hypoxic mDCs endowed with proinflammatory properties.


Subject(s)
Cell Differentiation/genetics , Dendritic Cells/cytology , Dendritic Cells/metabolism , Gene Expression Profiling , Membrane Glycoproteins/metabolism , Receptors, Immunologic/genetics , Adaptor Proteins, Signal Transducing/metabolism , Arthritis, Juvenile/genetics , Arthritis, Juvenile/pathology , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Chemokines/metabolism , Cross-Linking Reagents/pharmacology , Data Mining , Dendritic Cells/drug effects , Gene Expression Regulation/drug effects , Humans , Inflammation Mediators/metabolism , Membrane Glycoproteins/genetics , Membrane Proteins/metabolism , Monocytes/cytology , Oxygen/pharmacology , Phenotype , Receptors, Immunologic/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Synovial Fluid/drug effects , Synovial Fluid/metabolism , Triggering Receptor Expressed on Myeloid Cells-1
8.
J Immunol ; 183(5): 3004-13, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19657094

ABSTRACT

Axl, a prototypic member of the transmembrane tyrosine kinase receptor family, is known to regulate innate immunity. In this study, we show that Axl expression is induced by IFN-alpha during human dendritic cell (DC) differentiation from monocytes (IFN/DC) and that constitutively Axl-negative, IL-4-differentiated DC (IL-4/DC) can be induced to up-regulate Axl by IFN-alpha. This effect is inhibited by TLR-dependent maturation stimuli such as LPS, poly(I:C), TLR7/8 ligand, and CD40L. LPS-induced Axl down-regulation on the surface of human IFN-alpha-treated DC correlates with an increased proteolytic cleavage of Axl and with elevated levels of its soluble form. GM6001 and TAPI-1, general inhibitors of MMP and ADAM family proteases, restored Axl expression on the DC surface and diminished Axl shedding. Furthermore, stimulation of Axl by its ligand, Gas6, induced chemotaxis of human DC and rescued them from growth factor deprivation-induced apoptosis. Our study provides the first evidence that Gas6/Axl-mediated signaling regulates human DC activities, and identifies Gas6/Axl as a new DC chemotaxis pathway. This encourages one to explore whether dysregulation of this novel pathway in human DC biology is involved in autoimmunity characterized by high levels of IFN-alpha.


Subject(s)
Cell Movement/immunology , Dendritic Cells/immunology , Intercellular Signaling Peptides and Proteins/physiology , Interferon-alpha/physiology , Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Signal Transduction/immunology , Cell Differentiation/immunology , Cell Survival/immunology , Cells, Cultured , Chemotaxis, Leukocyte/immunology , Dendritic Cells/cytology , Dendritic Cells/metabolism , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Interleukin-4/physiology , Lipopolysaccharides/physiology , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/biosynthesis , Up-Regulation/immunology , Axl Receptor Tyrosine Kinase
SELECTION OF CITATIONS
SEARCH DETAIL
...