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










Publication year range
1.
J Neuroinflammation ; 17(1): 339, 2020 Nov 12.
Article in English | MEDLINE | ID: mdl-33183330

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) is a common pathological condition that presently lacks a specific pharmacological treatment. Adenosine levels rise following TBI, which is thought to be neuroprotective against secondary brain injury. Evidence from stroke and inflammatory disease models suggests that adenosine signaling through the G protein-coupled A3 adenosine receptor (A3AR) can provide antiinflammatory and neuroprotective effects. However, the role of A3AR in TBI has not been investigated. METHODS: Using the selective A3AR agonist, MRS5980, we evaluated the effects of A3AR activation on the pathological outcomes and cognitive function in CD1 male mouse models of TBI. RESULTS: When measured 24 h after controlled cortical impact (CCI) TBI, male mice treated with intraperitoneal injections of MRS5980 (1 mg/kg) had reduced secondary tissue injury and brain infarction than vehicle-treated mice with TBI. These effects were associated with attenuated neuroinflammation marked by reduced activation of nuclear factor of kappa light polypeptide gene enhancer in B cells (NFκB) and MAPK (p38 and extracellular signal-regulated kinase (ERK)) pathways and downstream NOD-like receptor pyrin domain-containing 3 inflammasome activation. MRS5980 also attenuated TBI-induced CD4+ and CD8+ T cell influx. Moreover, when measured 4-5 weeks after closed head weight-drop TBI, male mice treated with MRS5980 (1 mg/kg) performed significantly better in novel object-placement retention tests (NOPRT) and T maze trials than untreated mice with TBI without altered locomotor activity or increased anxiety. CONCLUSION: Our results provide support for the beneficial effects of small molecule A3AR agonists to mitigate secondary tissue injury and cognitive impairment following TBI.


Subject(s)
Adenosine A3 Receptor Agonists/administration & dosage , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Neurocognitive Disorders/drug therapy , Neurocognitive Disorders/metabolism , Receptor, Adenosine A3/metabolism , Animals , Brain Injuries, Traumatic/pathology , Drug Delivery Systems/methods , Male , Mice , Mice, Inbred C57BL , Neurocognitive Disorders/pathology
2.
Arch Pharm Res ; 43(5): 540-552, 2020 May.
Article in English | MEDLINE | ID: mdl-32430718

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is the leading cause of human death worldwide. Currently available therapies for COPD mainly relieve symptoms and preserve lung function, suggesting the need to develop novel therapeutic or preventive regimens. Because chronic inflammation is a mechanism of emphysematous lesion formation and because adenosine A3 receptor signaling and peroxisome proliferator-activated receptor gamma (PPARγ) regulate inflammation, we investigated the effect of LJ-529, a selective adenosine A3 receptor agonist and partial PPARγ agonist, on inflammation in vitro and elastase-induced pulmonary emphysema in vivo. LJ-529 markedly ameliorated elastase-induced emphysematous lesion formation in the lungs in vivo, as indicated by the restoration of pulmonary function, suppression of airspace enlargement, and downregulation of elastase-induced matrix metalloproteinase activity and apoptotic cell death in the lungs. LJ-529 induced the expression of PPARγ target genes, the activity of PPARγ and several cytokines involved in inhibiting inflammation and inducing anti-inflammatory M2-like phenotypes. Moreover, LJ-529 did not exhibit significant cytotoxicity in normal cell lines derived from various organs in vitro and induced minimal changes in body weight in vivo, suggesting no overt toxicity of LJ-529 in vitro or in vivo. These results indicate the potential of LJ-529 as a novel therapeutic/preventive agent for emphysema with limited toxicity.


Subject(s)
Adenosine A3 Receptor Agonists/pharmacology , Adenosine/analogs & derivatives , Anti-Inflammatory Agents/pharmacology , PPAR gamma/agonists , Pulmonary Emphysema/drug therapy , Receptor, Adenosine A3/metabolism , Thionucleosides/pharmacology , Adenosine/administration & dosage , Adenosine/pharmacology , Adenosine A3 Receptor Agonists/administration & dosage , Administration, Oral , Animals , Anti-Inflammatory Agents/administration & dosage , Cell Line , Cell Survival/drug effects , Humans , Mice , Mice, Inbred Strains , PPAR gamma/genetics , Pancreatic Elastase/antagonists & inhibitors , Pancreatic Elastase/metabolism , Pulmonary Emphysema/metabolism , Thionucleosides/administration & dosage
3.
J Cancer Res Ther ; 13(1): 107-112, 2017.
Article in English | MEDLINE | ID: mdl-28508842

ABSTRACT

AIM OF THE STUDY: The cell cycle, a vital process that involves in cells' growth and division, lies at the heart of cancer. It has been shown that IB-MECA, an A3 adenosine receptor agonist inhibits the proliferation of cancer cells by inducing cell cycle arrest in several tumors. In this study, we evaluated the role of IB-MECA inhibition in cell cycle progression in ovarian cancer cells. MATERIALS AND METHODS: Cell viability was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay in Caov-4 and OVCAR-3. Analysis of cell cycle distribution was carried out by flow cytometry. To determine the mechanisms of IB-MECA-mediated induction of cell cycle arrest, the expression of cell cycle regulatory proteins Cyclin D1 and cyclin-dependent kinase 4 (CDK4) was evaluated. RESULTS: Our results showed that IB-MECA significantly reduced cell viability in a dose-dependent manner. Moreover, our results indicated that a low concentration of IB-MECA induced G1 cell cycle arrest. Reduction of Cyclin D1 and CDK4 protein levels was also observed after treating cancer cells with IB-MECA. CONCLUSION: This study demonstrated that IB-MECA induces G1 phase cell cycle arrest through Cyclin D1/CDK4-mediated pathway in ovarian cancer cells.


Subject(s)
Adenosine A3 Receptor Agonists/administration & dosage , Adenosine/analogs & derivatives , Cyclin D/genetics , Cyclin-Dependent Kinase 4/genetics , Ovarian Neoplasms/drug therapy , Adenosine/administration & dosage , Cell Division/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Flow Cytometry , G1 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Receptor, Adenosine A3/genetics , Signal Transduction/drug effects
4.
Neuropharmacology ; 114: 101-113, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27914963

ABSTRACT

Small mammals have the ability to enter torpor, a hypothermic, hypometabolic state, allowing impressive energy conservation. Administration of adenosine or adenosine 5'-monophosphate (AMP) can trigger a hypothermic, torpor-like state. We investigated the mechanisms for hypothermia using telemetric monitoring of body temperature in wild type and receptor knock out (Adora1-/-, Adora3-/-) mice. Confirming prior data, stimulation of the A3 adenosine receptor (AR) induced hypothermia via peripheral mast cell degranulation, histamine release, and activation of central histamine H1 receptors. In contrast, A1AR agonists and AMP both acted centrally to cause hypothermia. Commonly used, selective A1AR agonists, including N6-cyclopentyladenosine (CPA), N6-cyclohexyladenosine (CHA), and MRS5474, caused hypothermia via both A1AR and A3AR when given intraperitoneally. Intracerebroventricular dosing, low peripheral doses of Cl-ENBA [(±)-5'-chloro-5'-deoxy-N6-endo-norbornyladenosine], or using Adora3-/- mice allowed selective stimulation of A1AR. AMP-stimulated hypothermia can occur independently of A1AR, A3AR, and mast cells. A1AR and A3AR agonists and AMP cause regulated hypothermia that was characterized by a drop in total energy expenditure, physical inactivity, and preference for cooler environmental temperatures, indicating a reduced body temperature set point. Neither A1AR nor A3AR was required for fasting-induced torpor. A1AR and A3AR agonists and AMP trigger regulated hypothermia via three distinct mechanisms.


Subject(s)
Adenosine A1 Receptor Agonists/administration & dosage , Adenosine A3 Receptor Agonists/administration & dosage , Adenosine Monophosphate/physiology , Fever/chemically induced , Receptor, Adenosine A1/physiology , Receptor, Adenosine A3/physiology , Torpor , Adenosine/administration & dosage , Adenosine/analogs & derivatives , Animals , Brain/drug effects , Brain/physiology , Histamine/metabolism , Injections, Intraventricular , Male , Mast Cells/drug effects , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Receptor, Adenosine A1/genetics , Receptor, Adenosine A3/genetics , Receptors, Histamine H1/physiology
5.
Mol Med Rep ; 14(5): 4335-4341, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27666664

ABSTRACT

Ischemia/reperfusion (IR) injury during clinical hepatic procedures is characterized by inflammatory conditions and the apoptosis of hepatocytes. Nuclear factor­κB (NF­κB), nitric oxide and the expression levels of inflammatory cytokines, tumor necrosis factor­α and interleukin­1 were observed to increase following IR and mediate the inflammatory response in the liver. CF102 is a highly selective A3 adenosine receptor (A3AR) agonist, and has been identified to induce an anti­inflammatory and protective effect on the liver via the downregulation of the NF­κB signaling pathway. The present study aimed to determine the effect of CF102 on protecting the liver against IR injury. The potential protective effect of CF102 (100 µg/kg) was assessed using an IR injury model on 70% of the liver of Wistar rats, which was induced by clamping the hepatic vasculature for 30 min. The regenerative effect of CF102 was assessed by the partial hepatectomy of 70% of the liver during 10 min of IR. CF102 reduced the levels of liver enzymes following IR injury. A higher regeneration rate in the CF102 treatment group was observed compared with the control group, suggesting that CF102 had a positive effect on the proliferation of hepatocytes following hepatectomy. CF102 had a protective effect on the liver of Wistar rats subsequent to IR injury during hepatectomy. This may be due to an anti­inflammatory and anti­apoptotic effect mediated by the A3AR.


Subject(s)
Adenosine A3 Receptor Agonists/administration & dosage , Adenosine/analogs & derivatives , Liver Regeneration/drug effects , Liver/drug effects , Receptor, Adenosine A3/metabolism , Reperfusion Injury/drug therapy , Adenosine/administration & dosage , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Hepatectomy , Hepatocytes/drug effects , Humans , Liver/injuries , Liver/pathology , Liver/surgery , Rats , Rats, Wistar , Receptor, Adenosine A3/genetics , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
6.
J Med Chem ; 59(7): 3249-63, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-26890707

ABSTRACT

Purine (N)-methanocarba-5'-N-alkyluronamidoriboside A3 adenosine receptor (A3AR) agonists lacking an exocyclic amine resulted from an unexpected reaction during a Sonogashira coupling and subsequent aminolysis. Because the initial C6-Me and C6-styryl derivatives had unexpectedly high A3AR affinity, other rigid nucleoside analogues lacking an exocyclic amine were prepared. Of these, the C6-Me-(2-phenylethynyl) and C2-(5-chlorothienylethynyl) analogues were particularly potent, with human A3AR Ki values of 6 and 42 nM, respectively. Additionally, the C2-(5-chlorothienyl)-6-H analogue was potent and selective at A3AR (MRS7220, Ki 60 nM) and also completely reversed mouse sciatic nerve mechanoallodynia (in vivo, 3 µmol/kg, po). The lack of a C6 H-bond donor while maintaining A3AR affinity and efficacy could be rationalized by homology modeling and docking of these hypermodified nucleosides. The modeling suggests that a suitable combination of stabilizing features can partially compensate for the lack of an exocyclic amine, an otherwise important contributor to recognition in the A3AR binding site.


Subject(s)
Adenosine A3 Receptor Agonists/pharmacology , Carbohydrates/chemistry , Nucleosides/chemistry , Pain/drug therapy , Purines/chemistry , Receptor, Adenosine A3/chemistry , Adenosine A3 Receptor Agonists/administration & dosage , Adenosine A3 Receptor Agonists/chemistry , Administration, Oral , Animals , CHO Cells , Cricetinae , Cricetulus , HEK293 Cells , Humans , Mice , Models, Molecular , Structure-Activity Relationship
7.
Sci Rep ; 5: 9047, 2015 Mar 12.
Article in English | MEDLINE | ID: mdl-25762375

ABSTRACT

The role of the adenosine A3 receptor (A3AR) in experimental colitis is controversial. The A3AR agonist N(6)-(3-iodobenzyl)adenosine-5'-N-methyluronamide (IB-MECA) has been shown to have a clinical benefit, although studies in A3AR-deficient mice suggest a pro-inflammatory role. However, there are no studies on the effect of 2-Cl-IB-MECA and the molecular mechanism of action of A3AR in murine colitis models in vivo. Is it the same as that observed in vitro? The interaction between 2-CL-IB-MECA and A3AR in a murine colitis model and the signaling pathways associated with this interaction remain unclear. Here we demonstrate a role for the NF-κB signaling pathway and its effect on modifying the activity of proinflammatory factors in A3AR-mediated biological processes. Our results demonstrated that A3AR activation possessed marked effects on experimental colitis through the NF-κB signaling pathway.


Subject(s)
Adenosine A3 Receptor Agonists/pharmacology , Colitis/metabolism , NF-kappa B/metabolism , Receptor, Adenosine A3/metabolism , Signal Transduction/drug effects , Adenosine/administration & dosage , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A3 Receptor Agonists/administration & dosage , Animals , Colitis/chemically induced , Colitis/drug therapy , Colitis/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gene Expression , Inflammation Mediators/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Peroxidase/metabolism , Receptor, Adenosine A3/genetics
8.
Glia ; 62(2): 199-216, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24311446

ABSTRACT

Adenosine receptor activation is involved in myelination and in apoptotic pathways linked to neurodegenerative diseases. In this study, we investigated the effects of adenosine receptor activation in the viability of oligodendrocytes of the rat optic nerve. Selective activation of A3 receptors in pure cultures of oligodendrocytes caused concentration-dependent apoptotic and necrotic death which was preceded by oxidative stress and mitochondrial membrane depolarization. Oligodendrocyte apoptosis induced by A3 receptor activation was caspase-dependent and caspase-independent. In addition to dissociated cultures, incubation of optic nerves ex vivo with adenosine and the A3 receptor agonist 2-CI-IB-MECA(1-[2-Chloro-6-[[(3-iodophenyl)methyl]amino]-9H-purin-9-yl]-1-deoxy-N-methyl-b-D-ribofuranuronamide)-induced caspase-3 activation, oligodendrocyte damage, and myelin loss, effects which were prevented by the presence of caffeine and the A3 receptor antagonist MRS 1220 (N-[9-Chloro-2-(2-furanyl)[1,2,4]-triazolo [1,5-c]quinazolin-5-yl]benzene acetamide). Finally, ischemia-induced injury and functional loss to the optic nerve was attenuated by blocking A3 receptors. Together, these results indicate that adenosine may trigger oligodendrocyte death via activation of A3 receptors and suggest that this mechanism contributes to optic nerve and white matter ischemic damage.


Subject(s)
Adenosine A3 Receptor Agonists/administration & dosage , Adenosine A3 Receptor Agonists/pharmacology , Apoptosis , Oligodendroglia/metabolism , Optic Nerve/metabolism , Receptor, Adenosine A3/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cells, Cultured , Oligodendroglia/cytology , Rats , Rats, Sprague-Dawley
9.
Arch Pharm Res ; 36(9): 1126-32, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23625750

ABSTRACT

The role of adenosine A3 receptors and their distribution in the gastrointestinal tract have been widely investigated. Most of the reports discuss their role in intestinal inflammations. However, the role of adenosine A3 receptor agonist in pancreatitis has not been well established. The aim of this study is [corrected] to evaluate the effects of the adenosine A3 receptor agonist on the course of sodium taurocholate-induced experimental acute pancreatitis (EAP). The experiments were performed on 80 male Wistar rats, 58 of which survived, subdivided into 3 groups: C--control rats, I--EAP group, and II--EAP group treated with the adenosine A3 receptor agonist IB-MECA (1-deoxy-1-6[[(3-iodophenyl) methyl]amino]-9H-purin-9-yl)-N-methyl-B-D-ribofuronamide at a dose of 0.75 mg/kg b.w. i.p. at 48, 24, 12 and 1 h before and 1 h after the injection of 5% sodium taurocholate solution into the biliary-pancreatic duct. Serum for α-amylase and lipase determinations and tissue samples for morphological examinations were collected at 2, 6, and 24 h of the experiment. In the IB-MECA group, α-amylase activity was decreased with statistically high significance compared to group I. The activity of lipase was not significantly different among the experimental groups but higher than in the control group. The administration of IB-MECA attenuated the histological parameters of inflammation as compared to untreated animals. The use of A3 receptor agonist IB-MECA attenuates EAP. Our findings suggest that stimulation of adenosine A3 receptors plays a positive role in the sodium taurocholate-induced EAP in rats.


Subject(s)
Adenosine A3 Receptor Agonists/therapeutic use , Adenosine/analogs & derivatives , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Pancreas/drug effects , Pancreatitis, Acute Necrotizing/prevention & control , Adenosine/administration & dosage , Adenosine/therapeutic use , Adenosine A3 Receptor Agonists/administration & dosage , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Disease Models, Animal , Edema/etiology , Edema/prevention & control , Injections, Intraperitoneal , Lipase/metabolism , Male , Necrosis , Pancreas/immunology , Pancreas/metabolism , Pancreas/pathology , Pancreatic alpha-Amylases/blood , Pancreatitis, Acute Necrotizing/immunology , Pancreatitis, Acute Necrotizing/metabolism , Pancreatitis, Acute Necrotizing/pathology , Rats , Rats, Wistar , Receptor, Adenosine A3/chemistry , Receptor, Adenosine A3/metabolism , Taurocholic Acid , Time Factors
10.
Physiol Res ; 61(6): 649-54, 2012.
Article in English | MEDLINE | ID: mdl-23098658

ABSTRACT

In our previous studies, IB-MECA, an adenosine A(3) receptor agonist, was found to stimulate proliferation of hematopoietic progenitor and precursor cells in mice. This property of IB-MECA was considered to be responsible for its ability to support regeneration of suppressed hematopoiesis after irradiation with sublethal doses of γ-rays when the drug was given in a post-irradiation treatment regimen. This study was aimed at assessing the ability of IB-MECA to influence a 30-day survival of lethally irradiated mice. In a series of experiments, IB-MECA was administered following various lethal radiation doses in various numbers of drug doses and various administration routes. Though in some of these experiments a moderate increase in 30-day survival was observed in IB-MECA-treated mice, the differences in comparison with the controls were not significantly different. It can be inferred from these results and those of previous studies assessing the effects of IB-MECA after sublethal radiation doses that IB-MECA can probably influence only a substantially preserved hematopoiesis like that remaining after sublethal irradiation. Future studies should be aimed at evaluation of the abilities of IB-MECA to influence post-irradiation survival when administered as a part of combined treatment regimens.


Subject(s)
Adenosine A3 Receptor Agonists/pharmacology , Adenosine/analogs & derivatives , Radiation Injuries, Experimental/mortality , Receptor, Adenosine A3/metabolism , Adenosine/administration & dosage , Adenosine/pharmacology , Adenosine A3 Receptor Agonists/administration & dosage , Animals , Gamma Rays , Hematopoiesis/drug effects , Hematopoiesis/radiation effects , Male , Mice , Mice, Inbred Strains , Radiation Injuries, Experimental/metabolism
11.
Pharmacol Res ; 65(3): 338-46, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22154845

ABSTRACT

Adenosine released during myocardial ischemia mediates cardioprotective preconditioning. Multivalent drugs covalently bound to nanocarriers may differ greatly in chemical and biological properties from the corresponding monomeric agents. Here, we conjugated chemically functionalized nucleosides to poly(amidoamine) (PAMAM) dendrimeric polymers and investigated their effects in rat primary cardiac cell cultures and in the isolated heart. Three conjugates of A3 adenosine receptor (AR) agonists, chain-functionalized at the C2 or N6 position, were cardioprotective, with greater potency than monomeric agonist Cl-IB-MECA. Multivalent amide-linked MRS5216 was selective for A1 and A3ARs, and triazole-linked MRS5246 and MRS5539 (optionally containing fluorescent label) were A3AR-selective. The conjugates protected ischemic rat cardiomyocytes, an effect blocked by an A3AR antagonist MRS1523, and isolated hearts with significantly improved infarct size, rate of pressure product, and rate of contraction and relaxation. Thus, strategically derivatized nucleosides tethered to biocompatible polymeric carriers display enhanced cardioprotective potency via activation of A3AR on the cardiomyocyte surface.


Subject(s)
Adenosine A3 Receptor Agonists/chemistry , Adenosine A3 Receptor Agonists/therapeutic use , Cardiotonic Agents/chemistry , Cardiotonic Agents/therapeutic use , Dendrimers/chemistry , Heart/drug effects , Myocardial Ischemia/drug therapy , Myocytes, Cardiac/drug effects , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A3 Receptor Agonists/administration & dosage , Adenosine A3 Receptor Agonists/pharmacology , Animals , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/pharmacology , Cells, Cultured , Male , Myocardial Ischemia/pathology , Myocytes, Cardiac/pathology , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A3/metabolism
12.
Biomed Pharmacother ; 65(6): 427-31, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21719245

ABSTRACT

Mouse hematopoiesis, suppressed by a sublethal dose of ionizing radiation, was the target for combined therapy with a cyclooxygenase-2 (COX-2) inhibitor meloxicam and an adenosine A3 receptor agonist IB-MECA. The drugs were administered in an early postirradiation treatment regimen: meloxicam was given in a single dose 1hour after irradiation, IB-MECA in two doses 24 and 48hours after irradiation. Treatment-induced changes in several compartments of hematopoietic progenitor and precursor cells of the bone marrow were evaluated on day 3 after irradiation. Values of hematopoietic progenitor cells for granulocytes/macrophages and erythrocytes (GM-CFC and BFU-E, respectively), as well as those of proliferative granulocytic cells were found to be significantly higher in the mice treated with the drug combination in comparison to irradiated controls and attained the highest increase factors of 1.6, 1.6, and 2.6, respectively. The study emphasizes the significance of the combined treatment of suppressed hematopoiesis with more agents. Mechanisms of the action of the individual compounds of the studied drug combination and of their joint operation are discussed.


Subject(s)
Adenosine A3 Receptor Agonists/therapeutic use , Cyclooxygenase 2 Inhibitors/therapeutic use , Gamma Rays/adverse effects , Hematopoiesis/drug effects , Radiation Injuries, Experimental/drug therapy , Adenosine/administration & dosage , Adenosine/analogs & derivatives , Adenosine/therapeutic use , Adenosine A3 Receptor Agonists/administration & dosage , Animals , Cell Count , Crosses, Genetic , Cyclooxygenase 2 Inhibitors/administration & dosage , Drug Therapy, Combination , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/radiation effects , Granulocyte Colony-Stimulating Factor/blood , Granulocyte Precursor Cells/drug effects , Granulocyte Precursor Cells/radiation effects , Hematinics/administration & dosage , Hematinics/therapeutic use , Hematopoiesis/radiation effects , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/radiation effects , Male , Meloxicam , Mice , Mice, Inbred CBA , Radiation Injuries, Experimental/blood , Radiation Injuries, Experimental/pathology , Thiazines/administration & dosage , Thiazines/therapeutic use , Thiazoles/administration & dosage , Thiazoles/therapeutic use , Whole-Body Irradiation
SELECTION OF CITATIONS
SEARCH DETAIL
...