Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J Bone Miner Res ; 34(5): 923-938, 2019 05.
Article in English | MEDLINE | ID: mdl-30645771

ABSTRACT

Osteopenia and fragility fractures have been associated with human immunodeficiency virus (HIV) infection. Tenofovir, a common antiviral in HIV treatment, also leads to increases in bone catabolism markers and decreased BMD in children and young adults. In murine models and human cell lines, tenofovir inhibits adenosine triphosphate release and decreases extracellular adenosine levels. Adenosine and adenosine A2A receptor inhibit osteoclast formation, and increase local adenosine concentration with dipyridamole, an agent that blocks adenosine cellular uptake and stimulates new bone formation as well as bone morphogenic protein 2. We hypothesized that tenofovir regulates bone resorption by diminishing endogenous adenosine levels and questioned whether dipyridamole may be a useful treatment to counteract the deleterous bone effects of tenofovir. Primary murine osteoclasts were induced by M-CSF/RANKL, and the number of TRAP-positive-cells was studied after challenge with tenofovir alone or in combination with dipyridamole. Differentiation markers were studied by RT-PCR and MAPK/NFkB expression by Western blot. Male C57Bl/6 mice were treated as follows: saline 0.9% (control), tenofovir 75 mg/kg/day, dipyridamole 25 mg/kg/day, combination tenofovir/dipyridamole (n = 10, 4 weeks). Calcein/Alizarin Red-labeling of newly formed bone was used, and long bones were prepared for micro-computed tomography (µCT)/histology. Tenofovir produced a dose-dependent increase in osteoclast differentiation (EC50 = 44.5nM) that was reversed by dipyridamole (IC50 = 0.3 µM). Tenofovir increased cathepsin K and NFATc1 mRNA levels and dipyridamole reversed the effect. Dipyridamole reversed the effect of tenofovir on pERK1/2, pp38, and NFkB nuclear translocation. Mice treated with tenofovir lost nearly 10% of their body weight (p < 0.001). µCT revealed decreased BMD and altered trabecular bone in tenofovir-treated mice, reversed by dipyridamole. TRAP-staining showed increased osteoclasts in tenofovir-treated mice (p < 0.005), an effect reversed by dipyridamole. Similar results were obtained for cathepsin K and CD68. RANKL-positive cells were increased in tenofovir-treated mice, whereas osteoprotegerin-positive cells were decreased; both effects were reversed by dipyridamole. These results suggest that treatment with agents that increase local adenosine concentrations, like dipyridamole, might prevent bone loss following tenofovir treatment. © 2019 American Society for Bone and Mineral Research.


Subject(s)
Bone Resorption , Dipyridamole/pharmacology , MAP Kinase Signaling System/drug effects , Osteoclasts/metabolism , Osteogenesis/drug effects , Tenofovir/adverse effects , Adenosine/metabolism , Animals , Bone Morphogenetic Protein 2/metabolism , Bone Resorption/chemically induced , Bone Resorption/drug therapy , Bone Resorption/metabolism , Bone Resorption/pathology , Female , Macrophage Colony-Stimulating Factor/metabolism , Mice , Osteoclasts/pathology , RANK Ligand/metabolism , Receptor, Adenosine A2A/metabolism , Tenofovir/pharmacology
2.
FASEB J ; 32(7): 3487-3501, 2018 07.
Article in English | MEDLINE | ID: mdl-29394106

ABSTRACT

The axonal guidance proteins semaphorin (Sema)4D and Sema3A play important roles in communication between osteoclasts and osteoblasts. As stimulation of adenosine A2A receptors (A2AR) regulates both osteoclast and osteoblast function, we asked whether A2AR regulates both osteoclast and osteoblast expression of Semas. In vivo bone formation and Sema3A/PlexinA1/Neuropilin-1, Sema4D/PlexinB1 protein expression were studied in a murine model of wear particle-induced osteolysis. Osteoclast/osteoblast differentiation were studied in vitro as the number of tartrate-resistant acid phosphatase+/Alizarin Red+ cells after challenge with CGS21680 (A2AR agonist, 1 µM) or ZM241385 (A2AR antagonist, 1 µM), with or without Sema4D or Sema3A (10 ng/ml). Sema3A/PlexinA1/Neuropilin-1, Sema4D/PlexinB1, and receptor activator of NF-κB ligand/osteoprotegerin (RANKL/OPG) expression was studied by RT-PCR and Western blot. ß-Catenin activation and cytoskeleton changes were studied by fluorescence microscopy and Western blot. In mice with wear particles implanted over the calvaria, CGS21680 treatment increased bone formation in vivo, reduced Sema4D, and increased Sema3A expression compared with mice with wear particle-induced osteolysis treated with vehicle alone. During osteoclast differentiation, CGS21680 abrogated RANKL-induced Sema4D mRNA expression (1.3 ± 0.3- vs. 2.5 ± 0.1-fold change, P < 0.001, n = 4). PlexinA1, but not Neuropilin-1, mRNA was enhanced by CGS21680 treatment. CGS21680 enhanced Sema3A mRNA expression during osteoblast differentiation (8.7 ± 0.2-fold increase, P < 0.001, n = 4); PlexinB1 mRNA was increased 2-fold during osteoblast differentiation and was not altered by CGS21680. Similar changes were observed at the protein level. CGS21680 decreased RANKL, increased OPG, and increased total/nuclear ß-catenin expression in osteoblasts. Sema4D increased Ras homolog gene family, member A phosphorylation and focal adhesion kinase activation in osteoclast precursors, and CGS21680 abrogated these effects. In summary, A2AR activation diminishes secretion of Sema4D by osteoclasts, inhibits Sema4D-mediated osteoclast activation, and enhances secretion of Sema3A by osteoblasts, increasing osteoblast differentiation and diminishing inflammatory osteolysis.-Mediero, A., Wilder, T., Shah, L., Cronstein, B. N. Adenosine A2A receptor (A2AR) stimulation modulates expression of semaphorins 4D and 3A, regulators of bone homeostasis.


Subject(s)
Adenosine A2 Receptor Agonists/pharmacology , Osteogenesis , Osteolysis , Receptor, Adenosine A2A/metabolism , Semaphorin-3A/metabolism , Semaphorins/metabolism , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Cells, Cultured , Female , Homeostasis , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuropilin-1/genetics , Neuropilin-1/metabolism , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Semaphorin-3A/genetics , Semaphorins/genetics , beta Catenin/genetics , beta Catenin/metabolism
3.
PLoS One ; 12(11): e0188135, 2017.
Article in English | MEDLINE | ID: mdl-29145453

ABSTRACT

BACKGROUND: Fibrosing diseases are a leading cause of morbidity and mortality worldwide and, therefore, there is a need for safe and effective antifibrotic therapies. Adenosine, generated extracellularly by the dephosphorylation of adenine nucleotides, ligates specific receptors which play a critical role in development of hepatic and dermal fibrosis. Results of recent clinical trials indicate that tenofovir, a widely used antiviral agent, reverses hepatic fibrosis/cirrhosis in patients with chronic hepatitis B infection. Belonging to the class of acyclic nucleoside phosphonates, tenofovir is an analogue of AMP. We tested the hypothesis that tenofovir has direct antifibrotic effects in vivo by interfering with adenosine pathways of fibrosis using two distinct models of adenosine and A2AR-mediated fibrosis. METHODS: Thioacetamide (100mg/kg IP)-treated mice were treated with vehicle, or tenofovir (75mg/kg, SubQ) (n = 5-10). Bleomycin (0.25U, SubQ)-treated mice were treated with vehicle or tenofovir (75mg/kg, IP) (n = 5-10). Adenosine levels were determined by HPLC, and ATP release was quantitated as luciferase-dependent bioluminescence. Skin breaking strength was analysed and H&E and picrosirus red-stained slides were imaged. Pannexin-1expression was knocked down following retroviral-mediated expression of of Pannexin-1-specific or scrambled siRNA. RESULTS: Treatment of mice with tenofovir diminished adenosine release from the skin of bleomycin-treated mice and the liver of thioacetamide-treated mice, models of diffuse skin fibrosis and hepatic cirrhosis, respectively. More importantly, tenofovir treatment diminished skin and liver fibrosis in these models. Tenofovir diminished extracellular adenosine concentrations by inhibiting, in a dose-dependent fashion, cellular ATP release but not in cells lacking Pannexin-1. CONCLUSIONS: These studies suggest that tenofovir, a widely used antiviral agent, could be useful in the treatment of fibrosing diseases.


Subject(s)
Adenosine Triphosphate/metabolism , Adenosine/metabolism , Antiviral Agents/pharmacology , Connexins/antagonists & inhibitors , Disease Models, Animal , Liver Cirrhosis/prevention & control , Liver/drug effects , Nerve Tissue Proteins/antagonists & inhibitors , Skin Diseases/prevention & control , Skin/drug effects , Tenofovir/pharmacology , Animals , Connexins/physiology , Dose-Response Relationship, Drug , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/physiology , Skin/metabolism
4.
Am J Pathol ; 187(9): 1935-1944, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28667836

ABSTRACT

Adenosine A2a receptor (A2aR) stimulation promotes the synthesis of collagens I and III, and we have recently demonstrated that there is crosstalk between the A2aR and WNT/ß-catenin signaling pathway. In in vitro studies, A2aR signaling for collagen III expression was mediated by WNT/ß-catenin signaling in human dermal fibroblasts; we further verified whether the crosstalk between A2aR and Wnt/ß-catenin signaling was involved in diffuse dermal fibrosis in vivo. Wnt-signaling reporter mice (Tcf/Lef:H2B-GFP) were challenged with bleomycin and treated with the selective A2aR antagonist istradefylline (KW6002) or vehicle. Dermal fibrosis was quantitated and nuclear translocation of ß-catenin in fibroblasts was assessed by double-staining for Green fluorescent protein or dephosphorylated ß-catenin or ß-catenin phosphorylated at Ser552, and vimentin. KW6002 significantly reduced skin thickness, skinfold thickness, breaking tension, dermal hydroxyproline content, myofibroblast accumulation, and collagen alignment in bleomycin-induced dermal fibrosis. Also, there was increased expression of Tcf/Lef:H2B-GFP reporter in bleomycin-induced dermal fibrosis, an effect that was diminished by treatment with KW6002. Moreover, KW6002 significantly inhibited nuclear translocation of Tcf/Lef:H2B-GFP reporter, as well as dephosphorylated ß-catenin and ß-catenin phosphorylated at Ser552. Our work supports the hypothesis that pharmacologic blockade of A2aR inhibits the WNT/ß-catenin signaling pathway, contributing to its capacity to inhibit dermal fibrosis in diseases such as scleroderma.


Subject(s)
Adenosine A2 Receptor Antagonists/pharmacology , Fibrosis/metabolism , Skin Diseases/metabolism , Skin/drug effects , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Animals , Bleomycin , Disease Models, Animal , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibrosis/chemically induced , Fibrosis/pathology , Mice , Phosphorylation/drug effects , Purines/pharmacology , Skin/metabolism , Skin/pathology , Skin Diseases/chemically induced , Skin Diseases/pathology
5.
Nat Commun ; 8: 15019, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28492224

ABSTRACT

Osteoarthritis (OA) is characterized by cartilage destruction and chondrocytes have a central role in this process. With age and inflammation chondrocytes have reduced capacity to synthesize and maintain ATP, a molecule important for cartilage homeostasis. Here we show that concentrations of ATP and adenosine, its metabolite, fall after treatment of mouse chondrocytes and rat tibia explants with IL-1ß, an inflammatory mediator thought to participate in OA pathogenesis. Mice lacking A2A adenosine receptor (A2AR) or ecto-5'nucleotidase (an enzyme that converts extracellular AMP to adenosine) develop spontaneous OA and chondrocytes lacking A2AR develop an 'OA phenotype' with increased expression of Mmp13 and Col10a1. Adenosine replacement by intra-articular injection of liposomal suspensions containing adenosine prevents development of OA in rats. These results support the hypothesis that maintaining extracellular adenosine levels is an important homeostatic mechanism, loss of which contributes to the development of OA; targeting adenosine A2A receptors might treat or prevent OA.


Subject(s)
5'-Nucleotidase/genetics , Adenosine/pharmacology , Arthritis, Experimental/drug therapy , Cartilage, Articular/drug effects , Chondrocytes/drug effects , Receptor, Adenosine A2A/genetics , 5'-Nucleotidase/deficiency , Adenosine/metabolism , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Chondrocytes/metabolism , Chondrocytes/pathology , Collagen Type X/genetics , Collagen Type X/metabolism , Gene Expression Regulation , Homeostasis , Humans , Injections, Intra-Articular , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/pharmacology , Liposomes/administration & dosage , Male , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoarthritis/drug therapy , Osteoarthritis/genetics , Osteoarthritis/metabolism , Osteoarthritis/pathology , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A2A/deficiency , Signal Transduction , Tibia/drug effects , Tibia/metabolism , Tibia/pathology
6.
J Biomed Mater Res B Appl Biomater ; 105(2): 366-375, 2017 02.
Article in English | MEDLINE | ID: mdl-26513656

ABSTRACT

Bone defects resulting from trauma or infection need timely and effective treatments to restore damaged bone. Using specialized three-dimensional (3D) printing technology we have created custom 3D scaffolds of hydroxyapatite (HA)/beta-tri-calcium phosphate (ß-TCP) to promote bone repair. To further enhance bone regeneration we have coated the scaffolds with dipyridamole, an agent that increases local adenosine levels by blocking cellular uptake of adenosine. Nearly 15% HA:85% ß-TCP scaffolds were designed using Robocad software, fabricated using a 3D Robocasting system, and sintered at 1100°C for 4 h. Scaffolds were coated with BMP-2 (200 ng mL-1 ), dypiridamole 100 µM or saline and implanted in C57B6 and adenosine A2A receptor knockout (A2AKO) mice with 3 mm cranial critical bone defects for 2-8 weeks. Dipyridamole release from scaffold was assayed spectrophotometrically. MicroCT and histological analysis were performed. Micro-computed tomography (microCT) showed significant bone formation and remodeling in HA/ß-TCP-dipyridamole and HA/ß-TCP-BMP-2 scaffolds when compared to scaffolds immersed in vehicle at 2, 4, and 8 weeks (n = 5 per group; p ≤ 0.05, p ≤ 0.05, and p ≤ 0.01, respectively). Histological analysis showed increased bone formation and a trend toward increased remodeling in HA/ß-TCP- dipyridamole and HA/ß-TCP-BMP-2 scaffolds. Coating scaffolds with dipyridamole did not enhance bone regeneration in A2AKO mice. In conclusion, scaffolds printed with HA/ß-TCP promote bone regeneration in critical bone defects and coating these scaffolds with agents that stimulate A2A receptors and growth factors can further enhance bone regeneration. These coated scaffolds may be very useful for treating critical bone defects due to trauma, infection or other causes. © 2015 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 366-375, 2017.


Subject(s)
Bone Morphogenetic Protein 2 , Bone Regeneration/drug effects , Calcium Phosphates , Coated Materials, Biocompatible , Dipyridamole , Durapatite , Printing, Three-Dimensional , Skull , Tissue Scaffolds/chemistry , Animals , Bone Morphogenetic Protein 2/chemistry , Bone Morphogenetic Protein 2/pharmacology , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Dipyridamole/chemistry , Dipyridamole/pharmacology , Durapatite/chemistry , Durapatite/pharmacology , Mice , Mice, Knockout , Skull/injuries , Skull/metabolism , Skull/pathology
7.
FASEB J ; 30(11): 3887-3900, 2016 11.
Article in English | MEDLINE | ID: mdl-27511945

ABSTRACT

As many as 10% of bone fractures heal poorly, and large bone defects resulting from trauma, tumor, or infection may not heal without surgical intervention. Activation of adenosine A2A receptors (A2ARs) stimulates bone formation. Ticagrelor and dipyridamole inhibit platelet function by inhibiting P2Y12 receptors and platelet phosphodiesterase, respectively, but share the capacity to inhibit cellular uptake of adenosine and thereby increase extracellular adenosine levels. Because dipyridamole promotes bone regeneration by an A2AR-mediated mechanism we determined whether ticagrelor could regulate the cells involved in bone homeostasis and regeneration in a murine model and whether inhibition of P2Y12 or indirect A2AR activation via adenosine was involved. Ticagrelor, dipyridamole and the active metabolite of clopidogrel (CAM), an alternative P2Y12 antagonist, inhibited osteoclast differentiation and promoted osteoblast differentiation in vitro. A2AR blockade abrogated the effects of ticagrelor and dipyridamole on osteoclast and osteoblast differentiation whereas A2BR blockade abrogated the effects of CAM. Ticagrelor and CAM, when applied to a 3-dimentional printed resorbable calcium-triphosphate/hydroxyapatite scaffold implanted in a calvarial bone defect, promoted significantly more bone regeneration than the scaffold alone and as much bone regeneration as BMP-2, a growth factor currently used to promote bone regeneration. These results suggest novel approaches to targeting adenosine receptors in the promotion of bone regeneration.-Mediero, A., Wilder, T., Reddy, V. S. R., Cheng, Q., Tovar, N., Coelho, P. G., Witek, L., Whatling, C., Cronstein, B. N. Ticagrelor regulates osteoblast and osteoclast function and promotes bone formation in vivo via an adenosine-dependent mechanism.


Subject(s)
Adenosine/analogs & derivatives , Osteoblasts/drug effects , Osteoclasts/drug effects , Adenosine/metabolism , Adenosine/pharmacology , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Bone Regeneration/drug effects , Clopidogrel , Dipyridamole/pharmacology , Mice, Inbred C57BL , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteogenesis/drug effects , Platelet Activation/drug effects , Receptor, Adenosine A2A/metabolism , Ticagrelor , Ticlopidine/analogs & derivatives , Ticlopidine/pharmacology
8.
FASEB J ; 30(11): 3835-3844, 2016 11.
Article in English | MEDLINE | ID: mdl-27502509

ABSTRACT

Rheumatoid arthritis is an autoimmune disease that is characterized by chronic inflammation and destruction of joints. Netrin-1, a chemorepulsant, laminin-like matrix protein, promotes inflammation by preventing macrophage egress from inflamed sites and is required for osteoclast differentiation. We asked whether blockade of Netrin-1 or its receptors [Unc5b and DCC (deleted in colorectal carcinoma)] may be useful therapeutic targets for treatment of inflammatory arthritis. Arthritis was induced in 8-wk-old C57Bl/6 mice by intraperitoneal injection of K/BxN serum. Murine monoclonal antibodies against Netrin-1, Unc5b, or DCC (10 µg/mouse) were injected weekly for 4 wk (n = 10). Paw swelling and thickness were assessed and following euthanasia 2-4 wk after serum transfer, paws were prepared for micro-computed tomography and histology. Paw inflammation was maximal 2 wk after injection. Anti-Netrin-1 or anti-Unc5b, but not anti-DCC, antibodies significantly reduced paw inflammation (clinical score: 9.8 ± 0.8, 10.4 ± 0.9, and 13.5 ± 0.5, respectively vs 16 ± 0 for control; P < 0.001). Micro-computed tomography showed bony erosions in untreated or anti-DCC-treated mice, whereas there were no erosions in anti-Netrin-1/anti-Unc5b-treated-animals. Tartrate-resistant acid phosphatase staining demonstrated a marked decrease in osteoclasts in anti-Netrin-1/anti-Unc5b-treated animals. Immunofluorescence staining revealed a decrease in cathepsin K+ and CD68+ cells in anti-Netrin-1/anti-Unc5b-treated animals. Blockade of Netrin-1/Unc5b by monoclonal antibodies prevents bone destruction and reduces the severity of K/BxN serum transfer-induced arthritis. Netrin-1 may be a novel therapeutic target for treatment of inflammatory bone destruction.-Mediero, A., Wilder, T., Ramkhelawon, B., Moore, K. J., Cronstein, B. N. Netrin-1 and its receptor Unc5b are novel targets for the treatment of inflammatory arthritis.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Macrophages/drug effects , Nerve Growth Factors/metabolism , Osteoclasts/drug effects , Receptors, Cell Surface/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Antibodies, Monoclonal/therapeutic use , Arthritis, Rheumatoid/immunology , Disease Models, Animal , Inflammation/drug therapy , Inflammation/immunology , Macrophages/metabolism , Mice, Transgenic , Nerve Growth Factors/genetics , Netrin Receptors , Netrin-1 , Osteoclasts/metabolism , Tumor Suppressor Proteins/genetics
9.
Purinergic Signal ; 12(3): 537-47, 2016 09.
Article in English | MEDLINE | ID: mdl-27289226

ABSTRACT

Bone homeostasis is a finely regulated mechanism involving different molecular pathways including adenosine signaling. The aim of this study is to determine the bone phenotype of adenosine A2B receptor knockout (A2BRKO) mice and to measure their ability to form new bone. Moreover, we analyzed the functionality of osteoclasts and osteoblasts from A2BRKO mice. Microcomputed tomography (µCT) analysis revealed a decrease of bone substance, bone mineral density, and trabecular number in A2BRKO mice compared to the WT mice at the same age. We measured the new bone formation by injecting fluorescent markers: it was reduced in femur and tibia of A2BRKO mice compare to the WT. A2BRKO young mice have fewer osteoblasts and an increase of osteoclasts was measured in the hind limbs of young and adult mice. A2BRKO osteoclasts are also more active in vitro, showing an increase of pit formation in dentin discs. Surprisingly in mature osteoblasts from A2BRKO mice, we measured an increase of calcified matrix production, collagen deposition, and alkaline phosphatase activity. These results demonstrate that A2BR on osteoblasts and osteoclasts regulate bone homeostasis.


Subject(s)
Bone and Bones/metabolism , Homeostasis/physiology , Osteogenesis/physiology , Receptor, Adenosine A2B/metabolism , Animals , Bone and Bones/pathology , Cell Differentiation/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , X-Ray Microtomography
10.
Ann Rheum Dis ; 75(9): 1706-13, 2016 09.
Article in English | MEDLINE | ID: mdl-26452536

ABSTRACT

OBJECTIVE: Netrin-1 is a chemorepulsant and matrix protein expressed during and required for osteoclast differentiation, which also plays a role in inflammation by preventing macrophage egress. Because wear particle-induced osteolysis requires osteoclast-mediated destruction of bone, we hypothesised that blockade of Netrin-1 or Unc5b, a receptor for Netrin-1, may diminish this pathological condition. METHODS: C57BL/6 mice, 6-8 weeks old, had 3 mg of ultrahigh-molecular-weight polyethylene particles implanted over the calvaria and then received 10 µg of monoclonal antibodies for Netrin-1 or its receptors, Unc5b and deleted in colon cancer (DCC), injected intraperitoneally on a weekly basis. After 2 weeks, micro-computed tomography and histology analysis were performed. Netrin-1 expression was analysed in human tissue obtained following primary prosthesis implantation or after prosthesis revision for peri-implant osteolysis and aseptic implant loosening. RESULTS: Weekly injection of anti-Netrin-1 or anti-Unc5b-antibodies significantly reduced particle-induced bone pitting in calvaria exposed to wear particles (46±4% and 49±3% of control bone pitting, respectively, p<0.001) but anti-DCC antibody did not affect inflammatory osteolysis (80±7% of control bone pitting, p=ns). Anti-Netrin-1 or anti-Unc5b, but not anti-DCC, antibody treatment markedly reduced the inflammatory infiltrate and the number of tartrate resistance acid phosphatase (TRAP)-positive osteoclasts (7±1, 4±1 and 14±1 cells/high power field (hpf), respectively, vs 12±1 cells/hpf for control, p<0.001), with no significant changes in alkaline phosphatase-positive osteoblasts on bone-forming surfaces in any antibody-treated group. Netrin-1 immunostaining colocalised with CD68 staining for macrophages. The peri-implant tissues of patients undergoing prosthesis revision surgery showed an increase in Netrin-1 expression, whereas there was little Netrin-1 expression in soft tissues removed at the time of primary joint replacement. CONCLUSIONS: These results demonstrate a unique role for Netrin-1 in osteoclast biology and inflammation and may be a novel target for prevention/treatment of inflammatory osteolysis.


Subject(s)
Nerve Growth Factors/physiology , Osteoclasts/physiology , Osteolysis/metabolism , Tumor Suppressor Proteins/physiology , Animals , Bone and Bones/metabolism , Bone and Bones/pathology , Inflammation , Male , Mice , Mice, Inbred C57BL , Nerve Growth Factors/antagonists & inhibitors , Netrin Receptors , Netrin-1 , Osteolysis/chemically induced , Osteolysis/pathology , Osteolysis/prevention & control , Polyethylenes , Receptors, Cell Surface/administration & dosage , Receptors, Cell Surface/antagonists & inhibitors , Skull/metabolism , Skull/pathology , Tumor Suppressor Proteins/antagonists & inhibitors
11.
Arthritis Res Ther ; 17: 249, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26370839

ABSTRACT

INTRODUCTION: This work was undertaken to delineate intracellular signaling pathways for the PDE4 inhibitor apremilast and to examine interactions between apremilast, methotrexate and adenosine A2A receptors (A2AR). METHODS: After apremilast and LPS incubation, intracellular cAMP, TNF-α, IL-10, IL-6 and IL-1α were measured in the Raw264.7 monocytic murine cell line. PKA, Epac1/2 (signaling intermediates for cAMP) and A2AR knockdowns were performed by shRNA transfection and interactions with A2AR and A2BR, as well as with methotrexate were tested in vitro and in the murine air pouch model. Statistical differences were determined using one or two-way ANOVA or Student's t test. The alpha nominal level was set at 0.05 in all cases. A P value of < 0.05 was considered significant. RESULTS: In vitro, apremilast increased intracellular cAMP and inhibited TNF-α release (IC50=104nM) and the specific A2AR-agonist CGS21680 (1µM) increased apremilast potency (IC50=25nM). In this cell line, apremilast increased IL-10 production. PKA, Epac1 and Epac2 knockdowns prevented TNF-α inhibition and IL-10 stimulation by apremilast. In the murine air pouch model, both apremilast and MTX significantly inhibited leukocyte infiltration, while apremilast, but not MTX, significantly inhibited TNF-α release. The addition of MTX (1 mg/kg) to apremilast (5 mg/kg) yielded no more inhibition of leukocyte infiltration or TNF-α release than with apremilast alone. CONCLUSIONS: The immunoregulatory effects of apremilast appear to be mediated by cAMP through the downstream effectors PKA, Epac1, and Epac2. A2AR agonism potentiated TNF-α inhibition by apremilast, consistent with the cAMP-elevating effects of that receptor. Because the A2AR is also involved in the anti-inflammatory effects of MTX, the mechanism of action of both drugs involves cAMP-dependent pathways and is therefore partially overlapping in nature.


Subject(s)
Cyclic AMP/metabolism , Inflammation Mediators/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Thalidomide/analogs & derivatives , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Antirheumatic Agents/pharmacology , Blotting, Western , Cell Line , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytokines/metabolism , Gene Expression/drug effects , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Methotrexate/pharmacology , Mice , Phenethylamines/pharmacology , RNA Interference , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Thalidomide/pharmacology , Tumor Necrosis Factor-alpha/metabolism
12.
FASEB J ; 29(4): 1577-90, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25573752

ABSTRACT

Promoting bone regeneration and repair of bone defects is a need that has not been well met to date. We have previously found that adenosine, acting via A2A receptors (A2AR) promotes wound healing and inhibits inflammatory osteolysis and hypothesized that A2AR might be a novel target to promote bone regeneration. Therefore, we determined whether direct A2AR stimulation or increasing endogenous adenosine concentrations via purine transport blockade with dipyridamole regulates bone formation. We determined whether coverage of a 3 mm trephine defect in a mouse skull with a collagen scaffold soaked in saline, bone morphogenetic protein-2 (BMP-2; 200 ng), 1 µM CGS21680 (A2AR agonist, EC50 = 160 nM), or 1 µM dipyridamole (EC50 = 32 nM) promoted bone regeneration. Microcomputed tomography examination demonstrated that CGS21680 and dipyridamole markedly enhanced bone regeneration as well as BMP-2 8 wk after surgery (60 ± 2%, 79 ± 2%, and 75 ± 1% bone regeneration, respectively, vs. 32 ± 2% in control, P < 0.001). Blockade by a selective A2AR antagonist (ZM241385, 1 µM) or deletion of A2AR abrogated the effect of CGS21680 and dipyridamole on bone regeneration. Both CGS21680 and dipyridamole treatment increased alkaline phosphatase-positive osteoblasts and diminished tartrate resistance acid phosphatase-positive osteoclasts in the defects. In vivo imaging with a fluorescent dye for new bone formation revealed a strong fluorescent signal in treated animals that was equivalent to BMP-2. In conclusion, stimulation of A2AR by specific agonists or by increasing endogenous adenosine levels stimulates new bone formation as well as BMP-2 and represents a novel approach to stimulating bone regeneration.


Subject(s)
Adenosine A2 Receptor Agonists/pharmacology , Bone Morphogenetic Protein 2/metabolism , Bone Regeneration/drug effects , Bone Regeneration/physiology , Receptor, Adenosine A2A/metabolism , Adenosine/analogs & derivatives , Adenosine/metabolism , Adenosine/pharmacology , Animals , Cell Count , Dipyridamole/pharmacology , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/metabolism , Phenethylamines/pharmacology
13.
Arthritis Rheumatol ; 67(3): 849-55, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25533750

ABSTRACT

OBJECTIVE: Adenosine, acting at the A2A receptor, mediates the antiinflammatory effects of methotrexate (MTX) in models of inflammation. We previously reported that A2A receptor ligation diminishes wear particle-driven osteolysis. The aim of this study was to investigate whether MTX treatment could prevent bone resorption caused by inflammatory osteolysis. METHODS: C57BL/6 mice (6-8 weeks old) received intraperitoneal injections of 1 mg/kg MTX (n = 20) or 0.9% saline (n = 10), starting 2 weeks prior to surgical implantation of 3 mg of wear particles (ultrahigh molecular weight polyethylene [UHMWPE] particles). The MTX-treated mice received daily injections of vehicle or ZM241385 at the surgical site until they were killed, 14 days later. XenoLight RediJect Bone Probe 680 was injected intravenously, and fluorescence analysis of the calvaria using an IVIS imaging system was performed to assess bone formation. Micro-computed tomography (micro-CT) and immunostaining for osteoclast and osteoblast markers were performed. RESULTS: Implantation of wear particles induced bone pitting and thinning, as shown by micro-CT. MTX treatment markedly reduced osteolysis, and this effect was abrogated by treatment with the A2A receptor antagonist ZM241385. Implantation of UHMWPE reduced new bone formation, and MTX treatment restored new bone formation, an effect that was completely reversed by treatment with ZM241385. Histologic examination of particle-exposed calvariae demonstrated that MTX prevented accumulation of an inflammatory infiltrate at the site of particle implantation, increased the number of osteoblasts, and reduced the number of osteoclasts at the site of inflammation, an effect that was reversed by treatment with ZM241385. CONCLUSION: MTX reduces inflammatory osteolysis indirectly via stimulation of A2A receptor and may represent a novel approach to enhance orthopedic implant survival, delaying or eliminating the need for revision arthroplasty surgery.


Subject(s)
Methotrexate/therapeutic use , Osteoclasts/drug effects , Osteolysis/prevention & control , Receptor, Adenosine A2A/metabolism , Skull/drug effects , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Humans , Inflammation , Male , Methotrexate/pharmacology , Mice , Mice, Inbred C57BL , Osteoclasts/metabolism , Osteolysis/chemically induced , Osteolysis/drug therapy , Osteolysis/metabolism , Polyethylenes , Skull/metabolism
14.
Nat Commun ; 4: 2909, 2013.
Article in English | MEDLINE | ID: mdl-24352507

ABSTRACT

Inflammasome pathways are important in chronic diseases; however, it is not known how the signalling is sustained after initiation. Inflammasome activation is dependent on stimuli such as lipopolysaccharide (LPS) and ATP that provide two distinct signals resulting in rapid production of interleukin (IL)-1ß, with the lack of response to repeat stimulation. Here we report that adenosine is a key regulator of inflammasome activity, increasing the duration of the inflammatory response via the A(2A) receptor. Adenosine does not replace signals provided by stimuli such as LPS or ATP but sustains inflammasome activity via a cAMP/PKA/CREB/HIF-1α pathway. In the setting of the lack of IL-1ß responses after previous exposure to LPS, adenosine can supersede this tolerogenic state and drive IL-1ß production. These data reveal that inflammasome activity is sustained, after initial activation, by A(2A) receptor-mediated signalling.


Subject(s)
Adenosine/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammasomes/metabolism , Receptor, Adenosine A2A/metabolism , Adenosine/pharmacology , Adenosine Triphosphate/metabolism , Animals , Carrier Proteins/metabolism , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Inflammasomes/drug effects , Interleukin-1beta/metabolism , Lipopolysaccharides/metabolism , Lipopolysaccharides/pharmacology , Liver/drug effects , Liver/immunology , Liver/metabolism , Liver/pathology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Receptor, Adenosine A2A/genetics , Signal Transduction/immunology
15.
Am J Pathol ; 183(6): 1740-1746, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24266925

ABSTRACT

Adenosine has an important role in inflammation and tissue remodeling and promotes dermal fibrosis by adenosine receptor (A2AR) activation. Adenosine may be formed intracellularly from adenine nucleotides or extracellularly through sequential phosphohydrolysis of released ATP by nucleoside triphosphate diphosphohydrolase (CD39) and ecto-5'-nucleotidase (CD73). Because the role of these ecto-enzymes in fibrosis appears to be tissue specific, we determined whether these ectonucleotidases were directly involved in diffuse dermal fibrosis. Wild-type and mice globally deficient in CD39 knockout (CD39KO), CD73 (CD73KO), or both (CD39/CD73DKO) were challenged with bleomycin. Extracellular adenosine levels and dermal fibrosis were quantitated. Adenosine release from skin cultured ex vivo was increased in wild-type mice after bleomycin treatment but remained low in skin from CD39KO, CD73KO, or CD39/CD73DKO bleomycin-treated mice. Deletion of CD39 and/or CD73 decreased the collagen content, and prevented skin thickening and tensile strength increase after bleomycin challenge. Decreased dermal fibrotic features were associated with reduced expression of the profibrotic mediators, transforming growth factor-ß1 and connective tissue growth factor, and diminished myofibroblast population in CD39- and/or CD73-deficient mice. Our work supports the hypothesis that extracellular adenosine, generated in tandem by ecto-enzymes CD39 and CD73, promotes dermal fibrogenesis. We suggest that biochemical or biological inhibitors of CD39 and/or CD73 may hold promise in the treatment of dermal fibrosis in diseases such as scleroderma.


Subject(s)
5'-Nucleotidase , Adenosine , Antigens, CD , Apyrase , Dermis , Scleroderma, Systemic , 5'-Nucleotidase/genetics , 5'-Nucleotidase/metabolism , Adenosine/genetics , Adenosine/metabolism , Animals , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/pharmacology , Antigens, CD/genetics , Antigens, CD/metabolism , Apyrase/genetics , Apyrase/metabolism , Bleomycin/adverse effects , Bleomycin/pharmacology , Dermis/metabolism , Dermis/pathology , Fibrosis/chemically induced , Fibrosis/genetics , Fibrosis/metabolism , Fibrosis/pathology , Mice , Mice, Knockout , Scleroderma, Systemic/chemically induced , Scleroderma, Systemic/genetics , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
16.
Br J Pharmacol ; 170(6): 1167-76, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23962057

ABSTRACT

BACKGROUND AND PURPOSE: Adenosine may be generated by hydrolysis of extracellular nucleotides by ectonucleotidases, including ectonucleoside triphosphate diphosphohydrolase 1 (CD39), ecto-5'-nucleotidase (CD73), nucleotide pyrophosphatase phosphodiesterase 1 (NPP-1) and tissue non-specific alkaline phosphatase (TNAP). Previous work from our laboratory has uncovered a critical role for adenosine A1 receptors (A1 R) in osteoclastogenesis; blockade or deletion of these receptors diminishes osteoclast differentiation. Interestingly, selective A1 R agonists neither affect basal osteoclastogenesis nor do they reverse A1 R antagonist-mediated inhibition of osteoclastogenesis. In this study, we determined whether ectonucleotidase-mediated adenosine production was required for A1 R antagonist-mediated inhibition, and, when we saw no effect, determined whether A1 R was constitutively activated and the antagonist was acting as an inverse agonist to diminish osteoclast differentiation. EXPERIMENTAL APPROACH: Osteoclast formation derived from wild-type, CD39 knockout (KO), CD73 KO, NPP-1 KO and TNAP KO mice was examined by tartrate-resistant acid phosphatase staining of receptor activator of NF-κB ligand-macrophage colony-stimulating factor-stimulated osteoclasts and osteoclast gene expression (Ctsk, Acp5, MMP-9 and NFATc1). Intracellular cAMP concentration was determined by elisa. KEY RESULTS: Rolofylline inhibited osteoclast formation in a dose-dependent manner (IC50 = 20-70 nM) in mice lacking all four of these phosphatases, although baseline osteoclast formation was significantly less in precursors from CD73 KO mice. Rolofylline (1 µM) stimulates cAMP production in bone marrow macrophages by 10.23 ± 0.89-fold. CONCLUSIONS AND IMPLICATIONS: Based on these findings, we hypothesize that the A1 R is constitutively activated in osteoclast precursors, thereby diminishing basal AC activity, and that A1 R antagonists act as inverse agonists to release A1 R-mediated inhibition of basal AC activity and permit osteoclast differentiation. The constitutive activity of A1 R promotes osteoclast formation and down-regulation of this activity blocks osteoclast formation.


Subject(s)
Adenosine A1 Receptor Antagonists/pharmacology , Osteoclasts/drug effects , Xanthines/pharmacology , 5'-Nucleotidase/deficiency , 5'-Nucleotidase/genetics , Alkaline Phosphatase/deficiency , Alkaline Phosphatase/genetics , Animals , Antigens, CD/genetics , Apyrase/deficiency , Apyrase/genetics , Bone Marrow Cells , Cell Differentiation/drug effects , Cells, Cultured , Cyclic AMP/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoclasts/cytology , Phosphoric Diester Hydrolases/deficiency , Phosphoric Diester Hydrolases/genetics , Pyrophosphatases/deficiency , Pyrophosphatases/genetics
17.
FASEB J ; 27(9): 3446-54, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23682121

ABSTRACT

Multiple myeloma (MM) is characterized by osteolytic bone lesions with uncoupled bone remodeling. In this study, we examined the effects of adenosine and its receptors (A1R, A2AR, A2BR, and A3R) on osteoblast and osteoclast differentiation of cells derived from patients with MM and healthy control subjects. Mesenchymal stem cells and bone marrow-derived mononuclear cells were isolated from bone marrow and differentiated into osteoblasts and osteoclasts, respectively. A1R antagonist rolofylline and A2BR agonist BAY60-6583 inhibit osteoclast differentiation of cells from patients with MM in a dose-dependent manner, as shown by TRAP staining (IC50: 10 and ∼10 nM, respectively). BAY60-6583 and dipyridamole, a nucleoside transport inhibitor, stimulate osteoblast differentiation of cells from patients with MM, as measured by ALP activity at d 14 and Alizarin Red staining at d 21 (by 1.57±0.03- and 1.71±0.45-fold, respectively), which can be blocked by A2BR antagonist MRS1754. Consistently, real-time PCR showed a significant increase of mRNA of osteocalcin and osterix at d 14. The effect of adenosine and its receptors is consistent in patients with MM and healthy subjects, suggesting an intrinsic mechanism that is important in both MM bone metabolism and normal physiology. Furthermore, the effect of dipyridamole on osteoblast differentiation is diminished in both A2BR- and CD39-knockout mice. These results indicate that adenosine receptors may be useful targets for the treatment of MM-induced bone disease.


Subject(s)
Adenosine/metabolism , Bone and Bones/metabolism , Multiple Myeloma/metabolism , Osteoclasts/metabolism , Receptor, Adenosine A1/metabolism , Receptor, Adenosine A2A/metabolism , Receptor, Adenosine A2B/metabolism , Acetamides/pharmacology , Aminopyridines/pharmacology , Animals , Bone and Bones/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Dipyridamole/pharmacology , Female , Humans , Mice , Mice, Knockout , Osteocalcin/genetics , Osteoclasts/drug effects , Purinergic P1 Receptor Agonists/pharmacology , Purines/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Xanthines/pharmacology
18.
Sci Transl Med ; 4(135): 135ra65, 2012 May 23.
Article in English | MEDLINE | ID: mdl-22623741

ABSTRACT

Prosthesis loosening, associated with wear particle-induced inflammation and osteoclast-mediated bone destruction, is a common cause for joint implant failure, leading to revision surgery. Adenosine A(2A) receptors (A(2A)Rs) mediate potent anti-inflammatory effects in many tissues and prevent osteoclast differentiation. We tested the hypothesis that an A(2A)R agonist could reduce osteoclast-mediated bone resorption in a murine calvaria model of wear particle-induced bone resorption. C57BL/6 and A(2A)R knockout (A(2A)R KO) mice received ultrahigh-molecular weight polyethylene particles and were treated daily with either saline or the A(2A)R agonist CGS21680. After 2 weeks, micro-computed tomography of calvaria demonstrated that CGS21680 reduced particle-induced bone pitting and porosity in a dose-dependent manner, increasing cortical bone and bone volume compared to control mice. Histological examination demonstrated diminished inflammation after treatment with CGS21680. In A(2A)R KO mice, CGS21680 did not affect osteoclast-mediated bone resorption or inflammation. Levels of bone resorption markers receptor activator of nuclear factor κB (RANK), RANK ligand, cathepsin K, CD163, and osteopontin were reduced after CGS21680 treatment, together with a reduction in osteoclasts. Secretion of interleukin-1ß (IL-1ß) and tumor necrosis factor-α was significantly decreased, whereas IL-10 was markedly increased in bone by CGS21680. These results in mice suggest that site-specific delivery of an adenosine A(2A)R agonist could enhance implant survival, delaying or eliminating the need for revision arthroplastic surgery.


Subject(s)
Adenosine A2 Receptor Agonists/therapeutic use , Adenosine/analogs & derivatives , Osteolysis/prevention & control , Phenethylamines/therapeutic use , Prosthesis Failure/adverse effects , Receptor, Adenosine A2A/metabolism , Adenosine/therapeutic use , Animals , Cells, Cultured , Female , Humans , Inflammation/drug therapy , Inflammation/etiology , Mice , Mice, Inbred C57BL , Mice, Knockout
19.
Am J Pathol ; 180(2): 775-86, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22138579

ABSTRACT

Adenosine is generated in increased concentrations at sites of injury/hypoxia and mediates a variety of physiological and pharmacological effects via G protein-coupled receptors (A(1), A(2A), A(2B), and A(3)). Because all adenosine receptors are expressed on osteoclasts, we determined the role of A(2A) receptor in the regulation of osteoclast differentiation. Differentiation and bone resorption were studied as the macrophage colony-stimulating factor-1-receptor activator of NF-κB ligand formation of multinucleated tartrate-resistant acid phosphatase (TRAP)-positive cells from primary murine bone marrow-derived precursors. A(2A) receptor and osteoclast marker expression levels were studied by RT-PCR. Cytokine secretion was assayed by enzyme-linked immunosorbent assay. In vivo examination of A(2A) knockout (KO)/control bones was determined by TRAP staining, micro-computed tomography, and electron microscopy. The A(2A) receptor agonist, CGS21680, inhibited osteoclast differentiation and function (half maximal inhibitory concentration, 50 nmol/L), increased the percentage of immature osteoclast precursors, and decreased IL-1ß and tumor necrosis factor-α secretion, an effect that was reversed by the A(2A) antagonist, ZM241385. Cathepsin K and osteopontin mRNA expression increased in control and ZM241385-pretreated osteoclasts, and this was blocked by CGS21680. Micro-computed tomography of A(2A)KO mouse femurs showed a significantly decreased bone volume/trabecular bone volume ratio, decreased trabecular number, and increased trabecular space. A(2A)KO femurs showed an increased TRAP-positive osteoclast. Electron microscopy in A(2A)KO femurs showed marked osteoclast membrane folding and increased bone resorption. Thus, adenosine, acting via the A(2A) receptor, inhibits macrophage colony-stimulating factor-1-receptor activator of NF-κB ligand-stimulated osteoclast differentiation and may regulate bone turnover under conditions in which adenosine levels are elevated.


Subject(s)
Cell Differentiation/physiology , Osteoclasts/cytology , Receptor, Adenosine A2A/physiology , Absorptiometry, Photon , Acid Phosphatase/metabolism , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Bone Density/physiology , Bone Resorption/pathology , Cells, Cultured , Cytokines/metabolism , Female , Femur/physiology , Isoenzymes/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Mice , Mice, Knockout , Osteoclasts/metabolism , RANK Ligand/pharmacology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Recombinant Proteins , Tartrate-Resistant Acid Phosphatase
20.
Purinergic Signal ; 7(1): 47-56, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21484096

ABSTRACT

Psychological stress has long been associated with effects on immune function and disease. In particular, differential effects of acute and chronic stress on skin immunity occur in the rodent restraint stress model, with acute stress enhancing and chronic stress suppressing cutaneous hypersensitivity. Extracellular levels of adenosine are known to modulate diverse biological activities in the CNS and peripheral tissues and serve an important protective function against physiological stressors such as inflammation and ischemia. In this study, we utilized the restraint stress model and the skin sensitizer dinitrofluorobezene to test the hypothesis that perceived stress influences contact hypersensitivity through an adenosine A(1) receptor-mediated mechanism. We subjected hapten-sensitized A(1) receptor knockout (A1 KO) mice and their wild-type (WT) littermates to either acute (2.5 h) or chronic (5 h daily × 4 weeks) restraint stress, followed by hapten re-challenge of the pinna. Daily measurements of the resulting pinna swellings from each group were compared to reactions in non-stressed controls. In WT mice, pinna swelling was augmented in acutely stressed mice and suppressed in the chronically stressed group. In contrast, contact hypersensitivity responses in the A1 KO mice failed to be affected by either acute or chronic stress. Absence of the adenosine A(1) receptor did not affect levels of plasma corticosterone or urine catecholamines under these stressful conditions but did lead to reduced numbers of circulating neutrophil granulocytes compared to stressed WT animals. These results suggest that the adenosine A(1) receptor pathway plays a role in the process by which perceived psychological stress influences the contact hypersensitivity response.

SELECTION OF CITATIONS
SEARCH DETAIL
...