Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 21(22)2020 Nov 20.
Article in English | MEDLINE | ID: mdl-33233631

ABSTRACT

The purinergic signaling has an important role in regulating pancreatic exocrine secretion. The exocrine pancreas is also a site of one of the most serious cancer forms, the pancreatic ductal adenocarcinoma (PDAC). Here, we explore how the network of purinergic and adenosine receptors, as well as ecto-nucleotidases regulate normal pancreatic cells and various cells within the pancreatic tumor microenvironment. In particular, we focus on the P2X7 receptor, P2Y2 and P2Y12 receptors, as well as A2 receptors and ecto-nucleotidases CD39 and CD73. Recent studies indicate that targeting one or more of these candidates could present new therapeutic approaches to treat pancreatic cancer. In pancreatic cancer, as much as possible of normal pancreatic function should be preserved, and therefore physiology of purinergic signaling in pancreas needs to be considered.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Pancreatic Neoplasms/drug therapy , Signal Transduction/genetics , 5'-Nucleotidase/genetics , 5'-Nucleotidase/immunology , Animals , Apyrase/genetics , Apyrase/immunology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Clinical Trials as Topic , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Gene Expression Regulation, Neoplastic/immunology , Humans , Immunotherapy/methods , Pancreas/drug effects , Pancreas/immunology , Pancreas/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/drug effects , Pancreatic Stellate Cells/immunology , Pancreatic Stellate Cells/pathology , Receptors, Adenosine A2/genetics , Receptors, Adenosine A2/immunology , Receptors, Purinergic P2X7/genetics , Receptors, Purinergic P2X7/immunology , Receptors, Purinergic P2Y12/genetics , Receptors, Purinergic P2Y12/immunology , Receptors, Purinergic P2Y2/genetics , Receptors, Purinergic P2Y2/immunology , Signal Transduction/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
2.
Immunol Cell Biol ; 95(4): 333-339, 2017 04.
Article in English | MEDLINE | ID: mdl-28174424

ABSTRACT

Tumor cells use various ways to evade anti-tumor immune responses. Adenosine, a potent immunosuppressive metabolite, is often found elevated in the extracellular tumor microenvironment. Therefore, targeting adenosine-generating enzymes (CD39 and CD73) or adenosine receptors has emerged as a novel means to stimulate anti-tumor immunity. In particular, the A2 (A2a and A2b) adenosine receptors exhibit similar immunosuppressive and pro-angiogenic functions, yet have distinct biological roles in cancer. In this review, we describe the common and distinct biological consequences of A2a and A2b adenosine receptor signaling in cancer. We discuss recent pre-clinical studies and summarize the different mechanisms-of-action of adenosine-targeting drugs. We also review the rationale for combining inhibitors of the adenosine pathway with other anticancer therapies such immune checkpoint inhibitors, tumor vaccines, chemotherapy and adoptive T cell therapy.


Subject(s)
Adenosine A2 Receptor Antagonists/therapeutic use , Cancer Vaccines/immunology , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Receptors, Adenosine A2/metabolism , 5'-Nucleotidase/metabolism , Animals , Antibodies, Monoclonal/therapeutic use , Antigens, CD/metabolism , Antineoplastic Agents/therapeutic use , Apyrase/metabolism , Combined Modality Therapy , Costimulatory and Inhibitory T-Cell Receptors/immunology , Humans , Immunity , Molecular Targeted Therapy , Neoplasms/immunology , Neovascularization, Pathologic , Receptors, Adenosine A2/immunology , Tumor Microenvironment
3.
Immunobiology ; 221(1): 84-93, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26307000

ABSTRACT

Regulatory T cells that express CD39 (CD39+ Treg) exhibit specific immunomodulatory properties. Ectonucleotidase CD39 hydrolyses ATP and ADP. ATP is a ligand of the P2X7 receptor and induces the shedding of CD62L and apoptosis. However, the role of ATP in CD39+ Treg cells has not been defined. Furthermore, NAD can activate the P2X7 receptor via ADP-ribosyltransferase (ART) enzymes and cause cell depletion in murine models. We evaluated the expression and function of P2X7 and ART1 in CD39+ Treg and CD39- Treg cells in the presence or absence of ATP and NAD. We isolated peripheral blood mononuclear cells from healthy subjects and purified CD4+ T cells, CD4+ CD25+ T cells and CD4+ CD25+ CD39+ T cells. P2X7 and ART1 expression was assessed by flow cytometry and real-time PCR. Our results showed low P2X7 expression on CD39+ Treg cells and higher levels of ART1 expression in CD4+ CD39+ T cells than the other subtypes studied. Neither shedding of CD62L nor cell death of CD39+ Treg or CD39- Treg cells was observed by 1mM ATP or 60µM NAD. In contrast, P2Xs receptor-dependent proliferation with 300µM ATP, was inhibited by NAD in the different cell types analysed. The NAD proliferation-inhibition was increased with P2Xs and A2a agonist and was reversed with P2Xs and A2a antagonist, therefore NAD inhibits P2Xs-dependent proliferation and A2a activation. In conclusion, our results suggest that the altered function and expression of P2X7 and ART1 in the human CD39+ Treg or CD39- Treg cells could participate in the resistance against cell death induced by ATP or NAD.


Subject(s)
ADP Ribose Transferases/immunology , Adenosine Triphosphate/pharmacology , NAD/pharmacology , Receptors, Purinergic P2X7/immunology , T-Lymphocyte Subsets/drug effects , ADP Ribose Transferases/genetics , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Adolescent , Adult , Antigens, CD/genetics , Antigens, CD/immunology , Apyrase/genetics , Apyrase/immunology , Cell Death/drug effects , Cell Proliferation , Female , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Gene Expression Regulation , Humans , Immunophenotyping , L-Selectin/genetics , L-Selectin/immunology , Male , Phenethylamines/pharmacology , Primary Cell Culture , Receptors, Adenosine A2/genetics , Receptors, Adenosine A2/immunology , Receptors, Purinergic P2X7/genetics , Signal Transduction , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Triazines/pharmacology , Triazoles/pharmacology
4.
J Immunol ; 194(8): 3735-44, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25780038

ABSTRACT

Dendritic cells (DC) are one target for immune suppression by regulatory T cells (Treg), because their interaction results in reduced T cell stimulatory capacity and secretion of inhibitory cytokines in DC. We show that DC in the presence of Treg are more mobile as compared with cocultures with conventional CD4(+) T cells and form DC-Treg aggregates within 2 h of culture. The migration of DC was specifically directed toward Treg, as Treg, but not CD4(+) T cells, attracted DC in Boyden chambers. Treg deficient for the ectonucleotidase CD39 were unable to attract DC. Likewise, addition of antagonists for A2A adenosine receptors abolished the formation of DC-Treg clusters, indicating a role for adenosine in guiding DC-Treg interactions. Analysis of the signal transduction events in DC after contact to Treg revealed increased levels of cAMP, followed by activation of Epac1 and the GTPase Rap1. Subsequently activated Rap1 localized to the subcortical actin cytoskeleton in DC, providing a means by which directed locomotion of DC toward Treg is facilitated. In aggregate, these data show that Treg degrade ATP to adenosine via CD39, attracting DC by activating Epac1-Rap1-dependent pathways. As a consequence, DC-Treg clusters are formed and DC are rendered less stimulatory. This adenosine-mediated attraction of DC may therefore act as one mechanism by which Treg regulate the induction of immune responses by DC.


Subject(s)
Adenosine/immunology , Cell Movement/immunology , Guanine Nucleotide Exchange Factors/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , rap1 GTP-Binding Proteins/immunology , Actin Cytoskeleton/immunology , Adenosine Triphosphate/immunology , Animals , Antigens, CD/immunology , Apyrase/immunology , Cell Communication/immunology , Dendritic Cells , Mice , Receptors, Adenosine A2/immunology
5.
J Mol Med (Berl) ; 91(2): 147-55, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23334369

ABSTRACT

The recent approval by the FDA of cancer vaccines and drugs that blockade immunological negative regulators has further enhanced interest in promising approaches of the immunotherapy of cancer. However, the disappointingly short life extension has also underscored the need to better understand the mechanisms that prevent tumor rejection and survival even after the blockade of immunological negative regulators. Here, we describe the implications of the "metabolism-based" immunosuppressive mechanism, where the local tissue hypoxia-driven accumulation of extracellular adenosine triggers suppression via A2 adenosine receptors on the surface of activated immune cells. This molecular pathway is of critical importance in mechanisms of immunosuppression in inflamed and cancerous tissue microenvironments. The protection of tumors by tumor-generated extracellular adenosine and A2 adenosine receptors could be the misguided application of the normal tissue-protecting mechanism that limits excessive collateral damage to vital organs during the anti-pathogen immune response. The overview of the current state of the art regarding the immunosuppressive effects of extracellular adenosine is followed by a historical perspective of studies focused on the elucidation of the physiological negative regulators that protect tissues of vital organs from excessive collateral damage, but, as a trade-off, may also weaken the anti-pathogen effector functions and negate the attempts of anti-tumor immune cells to destroy cancerous cells.


Subject(s)
Adenosine/immunology , Hypoxia/immunology , Immunotherapy, Adoptive , Neoplasms/therapy , Animals , Humans , Neoplasms/immunology , Receptors, Adenosine A2/immunology , Signal Transduction , T-Lymphocytes/immunology , Tumor Microenvironment
6.
Hum Immunol ; 72(7): 547-52, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21513753

ABSTRACT

Space flight and gravitational stress can alter innate immune function. Parabolic flights (PFs) as a model for short-term gravitational changes prime the cytotoxic capability of polymorphonuclear leukocytes (PMNs). In view of the emerging role of adenosine in the regulation of innate immune responses, we examined the potency of adenosine to control the release of cytotoxic H(2)O(2) by primed PMNs via the adenosine receptor system. During PFs, microgravity conditions (<10(-2) G) are generated for approximately 22 seconds, followed by a hypergravity (1.8 G) phase resulting in gravitational stress. We studied the ex vivo effects of adenosine on the production of H(2)O(2) by stimulated PMNs and determined adenosine plasma levels and adenosine A2(A) receptor transcripts of leukocytes of PF participants (n = 15). Increasing concentrations of adenosine dose dependently reduced tissue-toxic H(2)O(2) production by PMNs with a half-maximal inhibitory concentration (IC(50)) of 19.5 nM before takeoff and 7.6 nM at 48 hours after PF. This increase in the adenosine-mediated inhibition of PMNs' H(2)O(2) production was completely reversed by addition of the A2(A) receptor antagonist ZM241385. PF induced a nonsignificant elevation in adenosine plasma levels; A2(A) receptor mRNA from leukocytes remained almost unchanged. Adenosine limits the oxidative stress response of PMNs after PFs through an upregulation of the adenosine A2(A) receptor function. This stop signal on inflammation is stronger than that under normal physiologic states and may limit further cytotoxic damage. Pharmacologic manipulation of the adenosine A2(A) receptor pathway could be a potential target for control of unwanted exacerbations of cytotoxic PMN functions.


Subject(s)
Adenosine/pharmacology , Gravity, Altered , Neutrophils/drug effects , Oxidative Stress/physiology , Receptors, Adenosine A2/immunology , Vasodilator Agents/pharmacology , Adenosine/blood , Adenosine A2 Receptor Antagonists/pharmacology , Adult , Gene Expression Regulation/immunology , Gravity, Altered/adverse effects , Humans , Hydrogen Peroxide/metabolism , Immunologic Factors/metabolism , Immunologic Factors/pharmacology , Male , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/immunology , Oxidative Stress/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Adenosine A2/genetics , Triazines/pharmacology , Triazoles/pharmacology , Tumor Necrosis Factor-alpha/metabolism
7.
Folia Microbiol (Praha) ; 54(4): 364-8, 2009.
Article in English | MEDLINE | ID: mdl-19826926

ABSTRACT

Adenosines, endogenous purine nucleosides, appear in the extracellular space under metabolically stressful conditions associated with ischemia, inflammation, and cell damage. Their activity on innate immunity is prevalently inhibitory and can develop both in infectious and neoplastic diseases. During cancer development, tumor cells that release high concentrations of adenosines can impair the function of tumor-infiltrating lymphocytes and assist tumor growth by neo-angiogenesis. We evaluated the influence of A(2) adenosine receptor (A(2)AR) agonist on cytotoxic-cell response comparing human with other mammalian species (rodents, pigs, goats), both in healthy and in cancer conditions. The A(2)AR agonist developed dose-dependent inhibition of the cytotoxic activity of immune effector cells in all studied species. However, variability of the response was observed in relation to the species and the target cells that were used. Altogether, our data indicate that the A(2)AR plays a central role in adenosine-mediated inhibition of immune response to tumors.


Subject(s)
Adenosine A2 Receptor Agonists , Adenosine/analogs & derivatives , Cytotoxicity, Immunologic/drug effects , Killer Cells, Natural/immunology , Adenosine/immunology , Adenosine/pharmacology , Animals , Cells, Cultured , Female , Goats , Humans , Killer Cells, Natural/drug effects , Male , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Rats , Rats, Wistar , Receptors, Adenosine A2/immunology , Rodentia , Swine
8.
J Leukoc Biol ; 83(3): 447-55, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18160539

ABSTRACT

Adenosine regulates the function of the innate and adaptive immune systems through targeting virtually every cell type that is involved in orchestrating an immune/inflammatory response. Of the four adenosine receptors (A(1), A(2A), A(2B), A(3)), A(2A) receptors have taken center stage as the primary anti-inflammatory effectors of extracellular adenosine. This broad, anti-inflammatory effect of A(2A) receptor activation is a result of the predominant expression of A(2A) receptors on monocytes/macrophages, dendritic cells, mast cells, neutrophils, endothelial cells, eosinophils, epithelial cells, as well as lymphocytes, NK cells, and NKT cells. A(2A) receptor activation inhibits early and late events occurring during an immune response, which include antigen presentation, costimulation, immune cell trafficking, immune cell proliferation, proinflammatory cytokine production, and cytotoxicity. In addition to limiting inflammation, A(2A) receptors participate in tissue remodeling and reparation. Consistent with their multifaceted, immunoregulatory action on immune cells, A(2A) receptors have been shown to impact the course of a wide spectrum of ischemic, autoimmune, infectious, and allergic diseases. Here, we review the regulatory roles of A(2A) receptors in immune/inflammatory diseases of various organs, including heart, lung, gut, liver, kidney, joints, and brain, as well as the role of A(2A) receptors in regulating multiple organ failure and sepsis.


Subject(s)
Inflammation/physiopathology , Receptors, Adenosine A2/physiology , Wounds and Injuries/physiopathology , Adenosine/physiology , Animals , Animals, Genetically Modified , Humans , Immunity , Models, Biological , Receptors, Adenosine A2/immunology , Receptors, Purinergic P1/physiology , Signal Transduction
9.
Immunol Res ; 36(1-3): 91-9, 2006.
Article in English | MEDLINE | ID: mdl-17337770

ABSTRACT

Adenosine suppresses the production of various cytokines/ chemokines and inhibits the cytotoxic activity of murine and human NK cells activated with IL-2 or Ly49D, NKp46-receptor crosslinking, respectively. These effects are mediated by the type A2A adenosine receptor via stimulation of adenylyl cyclase, increased production of cAMP, and activation of PKA. PKA I, but not PKA II, participates in the inhibitory effects of adenosine. Blocking regulatory, but not catalytic, subunits of PKA I abrogates the inhibitory effects of adenosine. These findings suggest that tumor-produced adenosine inhibits the activity of NK and other effector cells and thereby protects tumors from immune-mediated destruction.


Subject(s)
Adenosine/immunology , Cyclic AMP-Dependent Protein Kinases/immunology , Killer Cells, Natural/immunology , Receptors, Adenosine A2/immunology , Tumor Escape , Adenosine/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Humans , Interleukin-2/metabolism , Isoenzymes/immunology , Isoenzymes/metabolism , Killer Cells, Natural/metabolism , Membrane Glycoproteins/metabolism , Natural Cytotoxicity Triggering Receptor 1 , Neoplasms/immunology , Receptors, Adenosine A2/metabolism , Receptors, Immunologic/metabolism
10.
J Infect Dis ; 189(10): 1897-904, 2004 May 15.
Article in English | MEDLINE | ID: mdl-15122527

ABSTRACT

BACKGROUND: Sepsis is currently treated with antibiotics and various adjunctive therapies that are not very effective. METHODS: Mouse survival (4-5 days) and peritoneal and blood bacteria counts were determined after challenge with intraperitoneal lipopolysaccharide (LPS) or live Escherichia coli. RESULTS: The A(2A) adenosine receptor (AR) agonist 4-[3-[6-amino-9-(5-ethylcarbamoyl-3, 4-dihydroxy-tetrahydro-furan-2-yl)-9H-purin-2-yl]-prop-2-ynyl]-cyclohexanecarboxylic acid methyl ester (ATL146e; 0.05-50 mu g/kg) protected mice from challenge with LPS, and protection occurred when treatment was delayed up to 24 h after challenge. Deletion of the A (2A) AR gene, Adora2a, inhibited protection by ATL146e. A putative A (3)AR agonist, N(6)-3-iodobenzyladenosine-5'-N-methyluronamide (IB-MECA; 500 mu g/kg but not 5 or 50 mu g/kg) protected mice from challenge with LPS. The protective effects of both ATL146e and IB-MECA were counteracted by the A(2A) AR selective antagonist 4-(2-[7-amino-2-[2-furyl][1,2,4]triazolo[2,3-a][1,3,5]triazin-5-yl-amino]ethyl)-phenol. In the live E. coli model, treatment with ATL146e (50 mu g/kg initiated 8 h after infection) increased survival in mice treated with ceftriaxone (5 days) from 40% to 100%. Treatment with ATL146e did not affect peritoneal numbers of live E. coli at the time of death or 120 h after infection but did increase numbers of peritoneal neutrophils and decreased the number of live E. coli in blood. CONCLUSIONS: AR agonists increase mouse survival in endotoxemia and sepsis via A(2A) AR-mediated mechanisms and reduce the number of live bacteria in blood.


Subject(s)
Adenosine/analogs & derivatives , Cyclohexanecarboxylic Acids/pharmacology , Endotoxemia/immunology , Escherichia coli Infections/immunology , Escherichia coli/immunology , Purines/pharmacology , Receptors, Adenosine A2/immunology , Sepsis/immunology , Adenosine/pharmacology , Adenosine A2 Receptor Agonists , Animals , Disease Models, Animal , Female , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Survival Analysis
11.
Annu Rev Immunol ; 22: 657-82, 2004.
Article in English | MEDLINE | ID: mdl-15032592

ABSTRACT

Immune cell-mediated destruction of pathogens may result in excessive collateral damage to normal tissues, and the failure to control activated immune cells may cause immunopathologies. The search for physiological mechanisms that downregulate activated immune cells has revealed a critical role for extracellular adenosine and for immunosuppressive A2A adenosine receptors in protecting tissue from inflammatory damage. Tissue damage-associated deep hypoxia, hypoxia-inducible factors, and hypoxia-induced accumulation of adenosine may represent one of the most fundamental and immediate tissue-protecting mechanisms, with adenosine A2A receptors triggering "OFF" signals in activated immune cells. In these regulatory mechanisms, oxygen deprivation and extracellular adenosine accumulation serve as "reporters," while A2A adenosine receptors serve as "sensors" of excessive tissue damage. The A2A receptor-triggered generation of intracellular cAMP then inhibits activated immune cells in a delayed negative feedback manner to prevent additional tissue damage. Targeting A2A adenosine receptors may have important clinical applications.


Subject(s)
DNA-Binding Proteins/immunology , Inflammation/immunology , Nuclear Proteins/immunology , Receptors, Adenosine A2/immunology , Adenosine/immunology , Animals , Humans , Hypoxia/immunology , Hypoxia/physiopathology , Immunity, Cellular , Signal Transduction/immunology
12.
Eur J Immunol ; 33(11): 3127-35, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14579281

ABSTRACT

Endothelial cell (EC) expression of proteins such as hemoxygenase-1 or Bcl-xL is associated with enhanced survival of vascularized allo- or xenografts. These grafts are resistant to humoral rejection, a phenomenon called accommodation. In vitro, low concentrations of allo- and xenoreactive antibody (XNA) induce a cytoprotective phenotype in EC similar to that seen in accommodated grafts. In this study we examine whether adenosine plays a role in antibody-induced cytoprotection. Porcine EC were incubated with human anti-pig XNA or specific alloantibody. EC expressed Bcl-xL and were protected from TNF-mediated apoptosis. Bcl-xL expression was inhibited by an adenosine A2 receptor antagonist. Human anti-pig XNA were shown to bind and induce cyclic adenosine 3',5'-monophosphate (cAMP) generation through these receptors. This activity was abolished by depletion of anti-galalpha(1-3)gal-specific XNA. In contrast, alloantibody caused adenosine production. Protection from TNF-mediated apoptosis was also mediated through A2 receptor but involved additional non-cAMP-dependent signaling. This study indicates a molecular mechanism common to both antibody-mediated cytoprotection and ischemic preconditioning and suggests a potential therapeutic avenue based on adenosine for improving the outcome of transplanted grafts in those patients with pre-existing anti-graft antibody.


Subject(s)
Antibodies/immunology , Cryopreservation , Endothelial Cells/immunology , Receptors, Adenosine A2/metabolism , Signal Transduction/physiology , Adenosine/metabolism , Animals , Endothelial Cells/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Adenosine A2/immunology , Swine/immunology , Swine/physiology , Vascular Cell Adhesion Molecule-1/metabolism , bcl-X Protein
SELECTION OF CITATIONS
SEARCH DETAIL