Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
Int J Mol Sci ; 24(7)2023 Mar 25.
Article in English | MEDLINE | ID: mdl-37047188

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are key determinants of the immunosuppressive microenvironment in tumors. As ion channels play key roles in the physiology/pathophysiology of immune cells, we aimed at studying the ion channel repertoire in tumor-derived polymorphonuclear (PMN-MDSC) and monocytic (Mo-MDSC) MDSCs. Subcutaneous tumors in mice were induced by the Lewis lung carcinoma cell line (LLC). The presence of PMN-MDSC (CD11b+/Ly6G+) and Mo-MDSCs (CD11b+/Ly6C+) in the tumor tissue was confirmed using immunofluorescence microscopy and cells were identified as CD11b+/Ly6G+ PMN-MDSCs and CD11b+/Ly6C+/F4/80-/MHCII- Mo-MDSCs using flow cytometry and sorting. The majority of the myeloid cells infiltrating the LLC tumors were PMN-MDSC (~60%) as compared to ~10% being Mo-MDSCs. We showed that PMN- and Mo-MDSCs express the Hv1 H+ channel both at the mRNA and at the protein level and that the biophysical and pharmacological properties of the whole-cell currents recapitulate the hallmarks of Hv1 currents: ~40 mV shift in the activation threshold of the current per unit change in the extracellular pH, high H+ selectivity, and sensitivity to the Hv1 inhibitor ClGBI. As MDSCs exert immunosuppression mainly by producing reactive oxygen species which is coupled to Hv1-mediated H+ currents, Hv1 might be an attractive target for inhibition of MDSCs in tumors.


Subject(s)
Myeloid-Derived Suppressor Cells , Neoplasms , Animals , Mice , Cell Line , Monocytes , Myeloid Cells , Myeloid-Derived Suppressor Cells/metabolism , Neoplasms/metabolism , Tumor Microenvironment
2.
J Cell Mol Med ; 23(9): 6203-6214, 2019 09.
Article in English | MEDLINE | ID: mdl-31273921

ABSTRACT

Nicotinic acid (NA) activates hydroxycarboxylic acid receptor 2 (HCA2 ), and it is widely used in treating dyslipidaemias. Since its side effects include skin dryness, whereas its deficiency can be accompanied by dyssebacia, characterized by sebaceous gland enlargement, we asked if HCA2 is expressed on human sebocytes, and if NA influences sebocyte functions. By using human immortalized SZ95 sebocytes, we found that non-cytotoxic (≤100 µmol/L; MTT-assay) concentrations of NA had no effect on the homeostatic sebaceous lipogenesis (SLG; Nile Red), but normalized excessive, acne-mimicking SLG induced by several lipogenic agents (arachidonic acid, anandamide, linoleic acid + testosterone; Nile Red; 48-hr treatments). Moreover, it exerted significant anti-proliferative actions (CyQUANT-assay), and increased [Ca2+ ]IC (Fluo-4 AM-based Ca2+ -measurement). Although NA did not prevent the lipopolysaccharide-induced pro-inflammatory response (up-regulation [Q-PCR] and release [ELISA] of several pro-inflammatory cytokines) of the sebocytes, collectively, these data support the concept that NA may be effective in suppressing sebum production in vivo. While exploring the mechanism of the sebostatic actions, we found that sebocytes express HCA2 (Q-PCR, immunofluorescent labelling), siRNA-mediated silencing of which prevented the NA-induced Ca2+ -signal and the lipostatic action. Collectively, our data introduce NA, and HCA2 activators in general, as novel, potent and most likely safe sebostatic agents, with possible anti-acne potential.


Subject(s)
Acne Vulgaris/genetics , Adenylyl Cyclases/genetics , Lipogenesis/drug effects , Niacin/pharmacology , Sebaceous Glands/drug effects , Acne Vulgaris/chemically induced , Acne Vulgaris/pathology , Arachidonic Acid/pharmacology , Cell Line , Cytokines/metabolism , Dyslipidemias/drug therapy , Dyslipidemias/pathology , Humans , Lipogenesis/genetics , Niacin/adverse effects , Niacin/genetics , RNA, Small Interfering/genetics , Sebaceous Glands/pathology
3.
J Lipid Res ; 54(9): 2458-74, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23833249

ABSTRACT

All-trans retinoic acid (ATRA) has a key role in dendritic cells (DCs) and affects T cell subtype specification and gut homing. However, the identity of the permissive cell types and the required steps of conversion of vitamin A to biologically active ATRA bringing about retinoic acid receptor-regulated signaling remains elusive. Here we present that only a subset of murine and human DCs express the necessary enzymes, including RDH10, RALDH2, and transporter cellular retinoic acid binding protein (CRABP)2, to produce ATRA and efficient signaling. These permissive cell types include CD103(+) DCs, granulocyte-macrophage colony-stimulating factor, and interleukin-4-treated bone marrow-derived murine DCs and human monocyte-derived DCs (mo-DCs). Importantly, in addition to RDH10 and RALDH2, CRABP2 also appears to be regulated by the fatty acid-sensing nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ) and colocalize in human gut-associated lymphoid tissue DCs. In our model of human mo-DCs, all three proteins (RDH10, RALDH2, and CRABP2) appeared to be required for ATRA production induced by activation of PPARγ and therefore form a linear pathway. This now functionally validated PPARγ-regulated ATRA producing and signaling axis equips the cells with the capacity to convert precursors to active retinoids in response to receptor-activating fatty acids and is potentially amenable to intervention in diseases involving or affecting mucosal immunity.


Subject(s)
Alcohol Oxidoreductases/metabolism , Dendritic Cells/metabolism , PPAR gamma/metabolism , Receptors, Retinoic Acid/metabolism , Retinal Dehydrogenase/metabolism , Signal Transduction , Tretinoin/metabolism , Alcohol Oxidoreductases/deficiency , Alcohol Oxidoreductases/genetics , Aldehyde Dehydrogenase 1 Family , Animals , Dendritic Cells/cytology , Dendritic Cells/enzymology , GTP-Binding Proteins/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Intestines/cytology , Male , Mice , Monocytes/cytology , Natural Killer T-Cells/cytology , Natural Killer T-Cells/metabolism , Protein Glutamine gamma Glutamyltransferase 2 , Protein Transport , Receptors, Retinoic Acid/deficiency , Receptors, Retinoic Acid/genetics , Retinal Dehydrogenase/deficiency , Retinal Dehydrogenase/genetics , Transglutaminases/metabolism
4.
J Immunol ; 187(1): 240-7, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21632707

ABSTRACT

It is well established that dendritic cells (DCs) take up, process, and present lipid Ags in complex with CD1d molecules to invariant NKT cells. The lipid-activated transcription factor, peroxisome proliferator-activated receptor γ (PPARγ), has previously been shown to regulate CD1d expression in human monocyte-derived DCs, providing a link between lipid metabolism and lipid Ag presentation. We report that PPARγ regulates the expression of a lysosomal protease, cathepsin D (CatD), in human monocyte-derived DCs. Inhibition of CatD specifically reduced the expansion of invariant NKT cells and furthermore resulted in decreased maturation of saposins, a group of lipid transfer proteins required for lysosomal lipid Ag processing and loading. These results reveal a novel mechanism of lipid Ag presentation and identify CatD as a key component of this machinery and firmly place PPARγ as the transcriptional regulator linking lipid metabolism and lipid Ag processing.


Subject(s)
Antigen Presentation/immunology , Cathepsin D/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Lipoproteins/metabolism , PPAR gamma/physiology , Antigens, CD1d/immunology , Antigens, CD1d/metabolism , Cathepsin D/biosynthesis , Cathepsin D/physiology , Cells, Cultured , Coculture Techniques , Humans , Lipid Metabolism/immunology , Lipoproteins/immunology , Lysosomes/enzymology , Lysosomes/metabolism , Monocytes/immunology , Monocytes/metabolism , Natural Killer T-Cells/enzymology , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Saposins/metabolism , Saposins/physiology , Signal Transduction/immunology , Up-Regulation/immunology
5.
Pharmaceuticals (Basel) ; 16(5)2023 Apr 27.
Article in English | MEDLINE | ID: mdl-37242439

ABSTRACT

5-chloro-2-guanidinobenzimidazole (ClGBI), a small-molecule guanidine derivative, is a known effective inhibitor of the voltage-gated proton (H+) channel (HV1, Kd ≈ 26 µM) and is widely used both in ion channel research and functional biological assays. However, a comprehensive study of its ion channel selectivity determined by electrophysiological methods has not been published yet. The lack of selectivity may lead to incorrect conclusions regarding the role of hHv1 in physiological or pathophysiological responses in vitro and in vivo. We have found that ClGBI inhibits the proliferation of lymphocytes, which absolutely requires the functioning of the KV1.3 channel. We, therefore, tested ClGBI directly on hKV1.3 using a whole-cell patch clamp and found an inhibitory effect similar in magnitude to that seen on hHV1 (Kd ≈ 72 µM). We then further investigated ClGBI selectivity on the hKV1.1, hKV1.4-IR, hKV1.5, hKV10.1, hKV11.1, hKCa3.1, hNaV1.4, and hNaV1.5 channels. Our results show that, besides HV1 and KV1.3, all other off-target channels were inhibited by ClGBI, with Kd values ranging from 12 to 894 µM. Based on our comprehensive data, ClGBI has to be considered a non-selective hHV1 inhibitor; thus, experiments aiming at elucidating the significance of these channels in physiological responses have to be carefully evaluated.

6.
Lab Invest ; 92(3): 345-61, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22157720

ABSTRACT

Granulomatous inflammations, characterized by the presence of activated macrophages (MAs) forming epithelioid cell (EPC) clusters, are usually easy to recognize. However, in ambiguous cases the use of a MA marker that expresses selectively in EPCs may be needed. Here, we report that carboxypeptidase-M (CPM), a MA-differentiation marker, is preferentially induced in EPCs of all granuloma types studied, but not in resting MAs. As CPM is not expressed constitutively in MAs, this allows utilization of CPM-immunohistochemistry in diagnostics of minute granuloma detection when dense non-granulomatous MAs are also present. Despite this rule, hardly any detectable CPM was found in advanced/active tubercle caseous disease, albeit in early tuberculosis granuloma, MAs still expressed CPM. Indeed, in vitro both the CPM-protein and -mRNA became downregulated when MAs were infected with live mycobacteria. In vitro, MA-CPM transcript is neither induced remarkably by interferon-γ, known to cause classical MA activation, nor by IL-4, an alternative MA activator. Instead, CPM is selectively expressed in lipid-laden MAs, including the foam cells of atherosclerotic plaques, xanthomatous lesions and lipid pneumonias. By using serum, rich in lipids, and low-density lipoprotein (LDL) or VLDL, CPM upregulation could be reproduced in vitro in monocyte-derived MAs both at transcriptional and protein levels, and the increase is repressed under lipid-depleted conditions. The microarray analyses support the notion that CPM induction correlates with a robust progressive increase in CPM gene expression during monocyte to MA maturation and dendritic cell (DC) differentiation mediated by granulocyte-MA-colony-stimulating factor+IL-4. M-CSF alone also induced CPM. These results collectively indicate that CPM upregulation in MAs is preferentially associated with increased lipid uptake, and exposure to CSF, features of EPCs, also. Therefore, CPM-immunohistochemistry is useful for granuloma and foam MA detections in tissue sections. Furthermore, the present data offer CPM for the first time to be a novel marker and cellular player in lipid uptake and/or metabolism of MAs by promoting foam cell formation.


Subject(s)
Epithelioid Cells/enzymology , Foam Cells/enzymology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Granuloma/metabolism , Metalloendopeptidases/metabolism , Biomarkers/metabolism , Cell Differentiation , Dendritic Cells/cytology , Dendritic Cells/metabolism , GPI-Linked Proteins/metabolism , Humans , Immunohistochemistry , Interleukin-4/metabolism , Lipid Metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Transcriptional Activation , Up-Regulation
7.
Methods Mol Biol ; 1182: 105-19, 2014.
Article in English | MEDLINE | ID: mdl-25055905

ABSTRACT

MicroRNAs are involved in the regulation of various pathophysiological processes such as immune regulation and cancer. Next-generation sequencing methods enable us to monitor their presence in various types of samples but we need flexible methods for validating datasets generated by high-throughput methods. Here we describe the detailed protocols to be used with our MiRNA Primer Design Tool assay design system. The presented methods allow the flexible design of the oligonucleotides needed for the RT-qPCR detection of any variant of small regulatory RNA molecules from virtually any species. This method can be used to measure miRNA levels from formalin-fixed, paraffin-embedded (FFPE) samples and various body fluids. As an example, we show the results of the hsa-miR-515-3p, hsa-miR-325, and hsa-miR-155 quantification using a specific UPL probe (Universal Probe Library) and a stem-loop RT-qPCR assay. The small nucleolar RNA RNU43 is used as endogenous control for normalization of the results. Urine from healthy pregnant women and FFPE samples from patients diagnosed with colorectal cancer and treated with antibody-based anti-EGFR monotherapy were used as samples.


Subject(s)
Body Fluids/chemistry , Gene Expression Profiling/methods , Animals , High-Throughput Nucleotide Sequencing , Humans , MicroRNAs/analysis , Paraffin , Paraffin Embedding , Reverse Transcriptase Polymerase Chain Reaction
8.
PPAR Res ; 2008: 473804, 2008.
Article in English | MEDLINE | ID: mdl-19009040

ABSTRACT

Dendritic cells (DCs) can regulate all elements of the immune system, and therefore are an ideal target for vaccination. During the last two decades, as a result of extensive research, DCs became the primary target of antitumor vaccination as well. A critical issue of antitumor vaccination is the phenotype of the dendritic cell used. It has been recently shown that several nuclear hormone receptors, and amongst them the lipid-activated nuclear receptor and peroxisome proliferator-activated receptor gamma (PPARgamma), have important roles in effecting the immunophenotype of human dendritic cells. It regulates primarily lipid metabolism and via this it influences the immunophenotype of DCs by altering lipid antigen uptake, presentation, and also other immune functions. In this review, we summarize the principles of antitumor vaccination strategies and present our hypothesis on how PPARgamma-regulated processes might be involved and could be exploited in the design of vaccination strategies.

SELECTION OF CITATIONS
SEARCH DETAIL