Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 194
Filter
1.
J Biomol Struct Dyn ; 41(2): 377-385, 2023 02.
Article in English | MEDLINE | ID: mdl-34851227

ABSTRACT

Adenosine deaminase is a zinc+2 dependent key enzyme of purine metabolism which irreversibly converts adenosine to inosine and form ammonia. Overexpression of adenosine deaminase has been linked to a variety of pathophysiological conditions such as atherosclerosis, hypertension, and diabetes. In the case of a cell-mediated immune response, ADA is thought to be a marker, particularly in type II diabetes. Deoxycoformycin is the most potent ADA inhibitor that has been discovered so far, but it has several drawbacks, including being toxic and having poor pharmacokinetics. Taxifolin, a flavonoid derived from plants, was discovered to be a potent inhibitor of the human ADA (hADA) enzyme in the current study. Taxifolin bound at the active site of human ADA and showed fifty percent inhibition at a concentration of 400 µM against the enzyme. To better understand the interactions between taxifolin and human ADA, docking and molecular dynamic simulations were performed. In-silico studies using autodock revealed that taxifolin bound in the active site of human ADA with a binding energy of -7.4 kcal mol -1 and a theoretical Ki of 3.7 uM. Comparative analysis indicated that taxifolin and deoxycoformycin share a common binding space in the active site of human ADA and inhibit its catalytic activity similarly. The work emphasises the need of employing taxifolin as a lead chemical in order to produce a more precise and effective inhibitor of the human ADA enzyme with therapeutic potential.Communicated by Ramaswamy H. Sarma.


Subject(s)
Adenosine Deaminase , Diabetes Mellitus, Type 2 , Humans , Adenosine Deaminase/chemistry , Adenosine Deaminase/metabolism , Pentostatin/pharmacology , Adenosine Deaminase Inhibitors/pharmacology , Adenosine Deaminase Inhibitors/chemistry
2.
Mol Ther ; 30(3): 1163-1170, 2022 03 02.
Article in English | MEDLINE | ID: mdl-34563676

ABSTRACT

The adenosine deaminase inhibitor 2'-deoxycoformycin (pentostatin, Nipent) has been used since 1982 to treat leukemia and lymphoma, but its mode of action is still unknown. Pentostatin was reported to decrease methylation of cellular RNA. We discovered that RNA extracted from pentostatin-treated cells or mice has enhanced immunostimulating capacities. Accordingly, we demonstrated in mice that the anticancer activity of pentostatin required Toll-like receptor 3, the type I interferon receptor, and T cells. Upon systemic administration of pentostatin, type I interferon is produced locally in tumors, resulting in immune cell infiltration. We combined pentostatin with immune checkpoint inhibitors and observed synergistic anti-cancer activities. Our work identifies pentostatin as a new class of an anticancer immunostimulating drug that activates innate immunity within tumor tissues and synergizes with systemic T cell therapies.


Subject(s)
Antineoplastic Agents , Lymphoma , Animals , Antineoplastic Agents/therapeutic use , Mice , Pentostatin/pharmacology , Pentostatin/therapeutic use , RNA , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/therapeutic use
3.
Antiviral Res ; 187: 105018, 2021 03.
Article in English | MEDLINE | ID: mdl-33476709

ABSTRACT

MBX-2168 has a mechanism of action similar to that of acyclovir (ACV) and ganciclovir (GCV), but two unique steps differentiate this drug from ACV/GCV. First, MBX-2168 is, at least partially, phosphorylated by the endogenous cellular kinase TAOK3 to a monophosphate. The second involves the removal of a moiety at the 6-position of MBX-2168-MP by adenosine deaminase like protein-1 (ADAL-1). It has been previously demonstrated that co-incubation with pentostatin (dCF), an ADAL-1 inhibitor, antagonizes the anti-viral activity of MBX-2168. We therefore hypothesize that inhibiting ADAL-1 results in a reduction of active compound produced in virus-infected cells. To test this, we examined the effect dCF has on the conversion of MBX-2168 to a triphosphate in HSV-1 and HCMV-infected cells. Our results demonstrate incubation of MBX-2168 alone or with dCF in HCMV-infected cells resulted in 53.1 ± 0.7 and 39.4 ± 1.5 pmol triphosphate/106 cells at 120 h, respectively. Incubation of MBX-2168 alone or with dCF in Vero cells resulted in 12.8 ± 0.1 and 6.7 ± 0.7 pmol triphosphate/106 cells at 24 h, respectively. HSV-1-infected Vero cells demonstrated no statistical difference in triphosphate accumulation at 24 h (13.1 ± 0.3 pmol triphosphate/106 cells). As expected, incubation with dCF resulted in the accumulation of MBX-2168-MP in both HFF (9.8 ± 0.9 pmol MBX-2168-MP/106 cells at 120 h) and Vero cells (4.7 ± 0.3 pmol MBX-2168-MP/106 cells at 24 h) while no detectable levels of monophosphate were observed in cultures not incubated with dCF. We conclude that dCF antagonizes the anti-viral effect of MBX-2168 by inhibiting the production of triphosphate, the active compound.


Subject(s)
Antiviral Agents/antagonists & inhibitors , Antiviral Agents/pharmacology , Cyclopropanes/antagonists & inhibitors , Cytomegalovirus/drug effects , Guanine/analogs & derivatives , Herpesvirus 1, Human/drug effects , Pentostatin/pharmacology , Polyphosphates/metabolism , Acyclovir/pharmacology , Animals , Cell Line , Chlorocebus aethiops , Cyclopropanes/pharmacology , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/virology , Fibroblasts/virology , Foreskin/cytology , Ganciclovir/pharmacology , Guanine/antagonists & inhibitors , Guanine/pharmacology , Herpes Simplex/drug therapy , Herpes Simplex/virology , Host Microbial Interactions , Humans , Loss of Function Mutation , Male , Phosphorylation , Vero Cells , Virus Replication/drug effects
4.
Exp Parasitol ; 199: 47-51, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30825499

ABSTRACT

The aim of this study was to evaluate in vitro the efficacy of cordycepin and pentostatin (alone or combined) against Trypanosoma cruzi, as well as the therapeutic efficiency of protocols of cordycepin and pentostatin combinations in mice experimentally infected with T. cruzi. In vitro, the cordycepin (3'-deoxyadenosine) and pentostatin (deoxycoformycin) exerted potent trypanocidal effect against T. cruzi (Colombian strain), similarly to benznidazole, which is the reference drug. For epimastigotes, the lethal dose of cordycepin capable of killing 50% (LD50) and 20% (LD20) of the parasites was 0.072 and 0.031 mg/mL, respectively and for trypomastigotes was 0.047 and 0.015 mg/mL, respectively. The combined use of cordycepin and pentostatin resulted in a LD50 and LD20 for epimastigotes of 0.068 and 0.027 mg/mL, respectively, as well as 0.056 and 0.018 mg/mL for trypomastigotes, respectively. In vivo, the combined use of cordycepin and pentostatin did not show the expected curative effect, however it was able to control the parasitema in the peak period. In summary, the combination of cordycepin and pentostatin showed no curative effect in mice infected by T. cruzi, despite the in vitro reduction of epimastigotes and trypomastigotes.


Subject(s)
Antiprotozoal Agents/pharmacology , Chagas Disease/drug therapy , Deoxyadenosines/pharmacology , Pentostatin/pharmacology , Trypanosoma cruzi/drug effects , Analysis of Variance , Animals , Antiprotozoal Agents/adverse effects , Antiprotozoal Agents/therapeutic use , Chagas Disease/parasitology , Deoxyadenosines/therapeutic use , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , Heart/drug effects , Lethal Dose 50 , Mice , Myocardium/pathology , Neglected Diseases/drug therapy , Neglected Diseases/parasitology , Nifurtimox/adverse effects , Nifurtimox/therapeutic use , Nitroimidazoles/adverse effects , Nitroimidazoles/therapeutic use , Nonlinear Dynamics , Parasitemia/prevention & control , Pentostatin/therapeutic use , Random Allocation , Regression Analysis
5.
J Mol Cell Cardiol ; 128: 62-76, 2019 03.
Article in English | MEDLINE | ID: mdl-30641086

ABSTRACT

Vascular inflammation is an important factor in the pathophysiology of cardiovascular diseases, such as atherosclerosis. Changes in the extracellular nucleotide and in particular adenosine catabolism may alter a chronic inflammation and endothelial activation. This study aimed to evaluate the relation between vascular ecto-adenosine deaminase (eADA) activity and endothelial activation in humans and to analyze the effects of LPS-mediated inflammation on this activity as well as mechanisms of its increase. Moreover, we investigated a therapeutic potential of ADA inhibition by deoxycofromycin (dCF) for endothelial activation. We demonstrated a positive correlation of vascular eADA activity and ADA1 mRNA expression with endothelial activation parameters in humans with atherosclerosis. The activation of vascular eADA was also observed under LPS stimulation in vivo along with endothelial activation, an increase in markers of inflammation and alterations in the lipid profile of a rat model. Ex vivo and in vitro studies on human specimen demonstrated that at an early stage of vascular pathology, eADA activity originated from activated endothelial cells, while at later stages also from an inflammatory infiltrate. We proposed that LPS-stimulated increase in endothelial adenosine deaminase activity could be a result of IL-6/JAK/STAT pathway activation, since the lack of IL-6 in mice was associated with lower vascular and plasma eADA activities. Furthermore, the inhibitors of JAK/STAT pathway decreased LPS-stimulated adenosine deaminase activity in endothelial cells. We demonstrated that cell surface eADA activity could be additionally regulated by transcytosis pathways, as exocytosis inhibitors including lipid raft inhibitor, methyl-ß-cyclodextrin decreased LPS-induced eADA activity. This suggests that cholesterol-dependent protein externalization mediated by lipid rafts could be an important factor in the eADA increase. Moreover, endocytosis inhibitors and exocytosis activators increased this activity on the cell surface. Furthermore, the inhibition of adenosine deaminase in endothelial cells in vitro attenuated LPS-mediated IL-6 release and soluble ICAM-1 and VCAM-1 concentration in the incubation medium through the restoration of the extracellular adenosine pool and adenosine receptor-dependent pathways. This study demonstrated that the vascular endothelial eADA activity remains under control of inflammatory mediators acting through JAK/STAT pathway that could be further modified by dyslipidemic-dependent exocytosis and transcytosis pathways. Inhibition of eADA blocked endothelial activation suggesting a crucial role of this enzyme in the control of vascular inflammation. This supports the concept of eADA targeted vascular protection therapy.


Subject(s)
Adenosine Deaminase/genetics , Aorta/metabolism , Atherosclerosis/genetics , Inflammation/genetics , Adenosine/genetics , Animals , Aorta/drug effects , Aorta/pathology , Atherosclerosis/enzymology , Atherosclerosis/pathology , Cell Membrane/drug effects , Cholesterol/genetics , Cholesterol/metabolism , Endothelial Cells/enzymology , Exocytosis/drug effects , Gene Expression Regulation/genetics , Humans , Inflammation/enzymology , Inflammation/pathology , Intercellular Adhesion Molecule-1/genetics , Interleukin-6/genetics , Janus Kinases/genetics , Lipopolysaccharides/pharmacology , Metabolism/genetics , Mice , Pentostatin/pharmacology , Rats , STAT Transcription Factors/genetics , Vascular Cell Adhesion Molecule-1/genetics
6.
J Cell Mol Med ; 22(12): 5939-5954, 2018 12.
Article in English | MEDLINE | ID: mdl-30291675

ABSTRACT

The activity of a cell-surface ecto-adenosine deaminase (eADA) is markedly increased in the endothelial activation and vascular inflammation leading to decreased adenosine concentration and alterations in adenosine signalling. Depending on the specific pathway activated, extracellular purines mediate host cell response or regulate growth and cytotoxicity on tumour cells. The aim of this study was to test the effects of adenosine deaminase inhibition by 2'deoxycoformycin (dCF) on the breast cancer development. dCF treatment decreased a tumour growth and a final tumour mass in female BALB/c mice injected orthotopically with 4T1 cancer cells. dCF also counteracted cancer-induced endothelial dysfunction in orthotopic and intravenous 4T1 mouse breast cancer models. In turn, this low dCF dose had a minor effect on immune stimulation exerted by 4T1 cell implantation. In vitro studies revealed that dCF suppressed migration and invasion of 4T1 cells via A2a and A3 adenosine receptor activation as well as 4T1 cell adhesion and transmigration through the endothelial cell layer via A2a receptor stimulation. Similar effects of dCF were observed in human breast cancer cells. Moreover, dCF improved a barrier function of endothelial cells decreasing its permeability. This study highlights beneficial effects of adenosine deaminase inhibition on breast cancer development. The inhibition of adenosine deaminase activity by dCF reduced tumour size that was closely related to the decreased aggressiveness of tumour cells by adenosine receptor-dependent mechanisms and endothelial protection.


Subject(s)
Adenosine Deaminase Inhibitors/pharmacology , Disease Progression , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Receptors, Purinergic P1/metabolism , Adenosine Deaminase/metabolism , Animals , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/pathology , Extracellular Space/metabolism , Female , Humans , Mammary Neoplasms, Animal/blood , Mammary Neoplasms, Animal/blood supply , Mice, Inbred BALB C , Neoplasm Invasiveness , Nucleotides/blood , Pentostatin/pharmacology , Phenotype , Transendothelial and Transepithelial Migration/drug effects
7.
Biol Blood Marrow Transplant ; 24(2): 308-313, 2018 02.
Article in English | MEDLINE | ID: mdl-29032276

ABSTRACT

In a multicenter, prospective, phase II study we evaluated the safety and efficacy of pentostatin followed by donor lymphocyte infusion (DLI) in patients with low donor Tcell chimerism after allogeneic hematopoietic cell transplantation (HCT). Thirty-six patients with low donor blood CD3 chimerism were enrolled in this study. Thirty-five patients received a total of 41 DLIs after a dose of pentostatin, and 1 patient received pentostatin only. Median donor CD3 chimerism prompting the initiation of pentostatin and DLI was 28% (range, 5% to 47%). Responses (defined by increases in donor CD3 chimerism ≥10% maintained to day 56 post-DLI) were seen in 16 patients (44.4%) with a median rise in CD3 donor chimerism to 64% (range, 48% to 100%). There was a trend for better responses among 21 patients who received first treatment within 100 days after transplant (57% response rate) compared with15 patients who received first treatment more than 100 days after HCT (27% response rate, P = .07). Fourteen patients (39%) developed grades II to IV acute graft-versus-host disease (GVHD) at a median of 10 days (range, 0 to 83) after DLI. Ten patients (28%) developed extensive chronic GVHD. Seventeen patients (47%) developed new grade 4 cytopenias after DLI. There was no difference in relapse between nonresponders and responders. Twenty-eight patients (78%) died, most (n = 21) because of relapse. Five of 16 responders (31%) are alive, all disease-free, at a median of 60 months (range, 21 to 132) after DLI. Six of 20 nonresponders (30%) are alive at a median of 47 months (range, 16 to 100) after DLI, 3 in complete remission. Pentostatin and DLI had acceptable toxicity and appeared to increase low donor CD3 chimerism after HCT but had no impact on mortality.


Subject(s)
Chimerism , Hematopoietic Stem Cell Transplantation , Lymphocyte Transfusion/methods , Pentostatin/administration & dosage , Adult , Aged , CD3 Complex/analysis , Female , Hematologic Neoplasms/mortality , Hematologic Neoplasms/therapy , Humans , Lymphocyte Transfusion/adverse effects , Male , Middle Aged , Pentostatin/pharmacology , Survival Analysis , Treatment Outcome
8.
Cell Chem Biol ; 24(2): 171-181, 2017 Feb 16.
Article in English | MEDLINE | ID: mdl-28111097

ABSTRACT

Pentostatin (PTN, deoxycoformycin) and arabinofuranosyladenine (Ara-A, vidarabine) are purine nucleoside antibiotics used clinically to treat hematological cancers and human DNA virus infections, respectively. PTN has a 1,3-diazepine ring, and Ara-A is an adenosine analog with an intriguing epimerization at the C-2' hydroxyl group. However, the logic underlying the biosynthesis of these interesting molecules has long remained elusive. Here, we report that the biosynthesis of PTN and Ara-A employs an unusual protector-protégé strategy. To our surprise, we determined that a single gene cluster governs PTN and Ara-A biosynthesis via two independent pathways. Moreover, we verified that PenB functions as a reversible oxidoreductase for the final step of PTN. Remarkably, we provided the first direct biochemical evidence that PTN can protect Ara-A from deamination by selective inhibition of the host adenosine deaminase. These findings expand our knowledge of natural product biosynthesis and open the way for target-directed genome mining of Ara-A/PTN-related antibiotics.


Subject(s)
Anti-Bacterial Agents/biosynthesis , Enzyme Inhibitors/metabolism , Pentostatin/biosynthesis , Vidarabine/biosynthesis , Adenosine Deaminase/metabolism , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Base Sequence , Cluster Analysis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Pentostatin/chemistry , Pentostatin/pharmacology , Sequence Analysis, DNA , Streptomyces antibioticus/genetics , Vidarabine/chemistry , Vidarabine/pharmacology
9.
Proc Natl Acad Sci U S A ; 113(28): E4079-87, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27357689

ABSTRACT

Engineered tumor-targeted anthrax lethal toxin proteins have been shown to strongly suppress growth of solid tumors in mice. These toxins work through the native toxin receptors tumor endothelium marker-8 and capillary morphogenesis protein-2 (CMG2), which, in other contexts, have been described as markers of tumor endothelium. We found that neither receptor is required for tumor growth. We further demonstrate that tumor cells, which are resistant to the toxin when grown in vitro, become highly sensitive when implanted in mice. Using a range of tissue-specific loss-of-function and gain-of-function genetic models, we determined that this in vivo toxin sensitivity requires CMG2 expression on host-derived tumor endothelial cells. Notably, engineered toxins were shown to suppress the proliferation of isolated tumor endothelial cells. Finally, we demonstrate that administering an immunosuppressive regimen allows animals to receive multiple toxin dosages and thereby produces a strong and durable antitumor effect. The ability to give repeated doses of toxins, coupled with the specific targeting of tumor endothelial cells, suggests that our strategy should be efficacious for a wide range of solid tumors.


Subject(s)
Antigens, Bacterial/therapeutic use , Bacterial Toxins/therapeutic use , Biomarkers, Tumor/metabolism , Endothelial Cells/drug effects , Neoplasms/drug therapy , Receptors, Peptide/metabolism , Animals , Antigens, Bacterial/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols , Bacterial Toxins/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclophosphamide/pharmacology , Cyclophosphamide/therapeutic use , Drug Screening Assays, Antitumor , Lymphocytes/drug effects , Mice , Microfilament Proteins , Molecular Targeted Therapy , Neoplasms/genetics , Pentostatin/pharmacology , Pentostatin/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Receptors, Cell Surface
10.
Cardiovasc Res ; 112(2): 590-605, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-28513806

ABSTRACT

AIMS: Extracellular nucleotides and adenosine that are formed or degraded by membrane-bound ecto-enzymes could affect atherosclerosis by regulating the inflammation and thrombosis. This study aimed to evaluate a relation between ecto-enzymes that convert extracellular adenosine triphosphate to adenine dinucleotide phosphate, adenosine monophosphate, adenosine, and inosine on the surface of the vessel wall with the severity or progression of experimental and clinical atherosclerosis. Furthermore, we tested whether the inhibition of adenosine deaminase will block the development of experimental atherosclerosis. METHODS AND RESULTS: Vascular activities of ecto-nucleoside triphosphate diphosphohydrolase 1, ecto-5'-nucleotidase, and ecto-adenosine deaminase (eADA) were measured in aortas of apolipoprotein E-/- low density lipoprotein receptor (ApoE-/-LDLR-/-) and wild-type mice as well as in human aortas. Plaques were analysed in the entire aorta, aortic root, and brachiocephalic artery by Oil-Red O and Orcein Martius Scarlet Blue staining and vascular accumulation of macrophages. The cellular location of ecto-enzymes was analysed by immunofluorescence. The effect of eADA inhibition on atherosclerosis progression was studied by a 2-month deoxycoformycin treatment of ApoE-/-LDLR-/- mice. The vascular eADA activity prominently increased in ApoE-/-LDLR-/- mice when compared with wild type already at the age of 1 month and progressed along atherosclerosis development, reaching a 10-fold difference at 10 months. The activity of eADA correlated with atherosclerotic changes in human aortas. High abundance of eADA in atherosclerotic vessels originated from activated endothelial cells and macrophages. There were no changes in ecto-nucleoside triphosphate diphosphohydrolase 1 activity, whereas ecto-5'-nucleotidase was moderately decreased in ApoE-/-LDLR-/- mice. Deoxycoformycin treatment attenuated plaque development in aortic root and brachiocephalic artery of ApoE-/-LDLR-/- mice, suppressed vascular inflammation and improved endothelial function. CONCLUSIONS: This study highlights the importance of extracellular nucleotides and adenosine metabolism in the atherosclerotic vessel in both experimental and clinical setting. The increased eADA activity marks an early stage of atherosclerosis, contributes to its progression and could represent a novel target for therapy.


Subject(s)
Adenosine Deaminase/metabolism , Atherosclerosis/metabolism , Adenosine/metabolism , Adenosine Deaminase Inhibitors/therapeutic use , Animals , Aorta/enzymology , Aorta/metabolism , Apolipoproteins E/physiology , Atherosclerosis/drug therapy , Cells, Cultured , Disease Models, Animal , Fluorescent Antibody Technique , Humans , Male , Mice , Mice, Inbred C57BL , Pentostatin/pharmacology , Receptors, LDL/physiology
11.
Naunyn Schmiedebergs Arch Pharmacol ; 388(8): 853-68, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25877465

ABSTRACT

The A1 adenosine and M2 muscarinic receptors exert protective (including energy consumption limiting) effects in the heart. We investigated the influence of adenosine deaminase (ADA) inhibition on a representative energy consumption limiting function, the direct negative inotropic effect elicited by the A1 adenosinergic and M2 muscarinergic systems, in eu- and hyperthyroid atria. Furthermore, we compared the change in the interstitial adenosine level caused by ADA inhibition and nucleoside transport blockade, two well-established processes to stimulate the cell surface A1 adenosine receptors, in both thyroid states. A classical isolated organ technique was applied supplemented with the receptorial responsiveness method (RRM), a concentration estimating procedure. Via measuring the contractile force, the direct negative inotropic capacity of N(6)-cyclopentyladenosine, a selective A1 receptor agonist, and methacholine, a muscarinic receptor agonist, was determined on the left atria isolated from 8-day solvent- and thyroxine-treated guinea pigs in the presence and absence of 2'-deoxycoformycin, a selective ADA inhibitor, and NBTI, a selective nucleoside transporter inhibitor. We found that ADA inhibition (but not nucleoside transport blockade) increased the signal amplification of the A1 adenosinergic (but not M2 muscarinergic) system. This action of ADA inhibition developed in both thyroid states, but it was greater in hyperthyroidism. Nevertheless, ADA inhibition produced a smaller rise in the interstitial adenosine concentration than nucleoside transport blockade did in both thyroid states. Our results indicate that ADA inhibition, besides increasing the interstitial adenosine level, intensifies the atrial A1 adenosinergic function in another (thyroid hormone-sensitive) way, suggesting a new mechanism of action of ADA inhibition.


Subject(s)
Adenosine Deaminase/physiology , Heart Atria/physiopathology , Hyperthyroidism/physiopathology , Receptor, Adenosine A1/physiology , Receptor, Muscarinic M2/physiology , Adenosine/physiology , Adenosine Deaminase Inhibitors/pharmacology , Animals , Guinea Pigs , Heart Atria/drug effects , Male , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Pentostatin/pharmacology , Thyroxine/pharmacology
12.
J Pharmacol Sci ; 127(1): 53-6, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25704018

ABSTRACT

Cordyceps sinensis, a fungus that parasitizes on the larva of Lepidoptera, has been used as a valued traditional Chinese medicine. We investigated the effects of water extracts of Cordyceps sinensis (WECS), and particularly focused on its anticancer and antimetastatic actions. Based on in vitro studies, we report that WECS showed an anticancer action, and this action was antagonized by an adenosine A3 receptor antagonist. Moreover, this anticancer action of WECS was promoted by an adenosine deaminase inhibitor. These results suggest that one of the components of WECS with an anticancer action might be an adenosine or its derivatives. Therefore, we focused on cordycepin (3'-deoxyadenosine) as one of the active ingredients of WECS. According to our experiments, cordycepin showed an anticancer effect through the stimulation of adenosine A3 receptor, followed by glycogen synthase kinase (GSK)-3ß activation and cyclin D1 suppression. Cordycepin also showed an antimetastatic action through inhibiting platelet aggregation induced by cancer cells and suppressing the invasiveness of cancer cells via inhibiting the activity of matrix metalloproteinase (MMP)-2 and MMP-9, and accelerating the secretion of tissue inhibitor of metalloproteinase (TIMP)-1 and TIMP-2 from cancer cells. In conclusion, cordycepin, an active component of WECS, might be a candidate anticancer and antimetastatic agent.


Subject(s)
Cordyceps/chemistry , Deoxyadenosines/therapeutic use , Neoplasm Metastasis/drug therapy , Pentostatin/pharmacology , Phytotherapy , Plant Extracts/pharmacology , Adenosine A3 Receptor Agonists/pharmacology , Adenosine A3 Receptor Agonists/therapeutic use , Adenosine A3 Receptor Antagonists/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Atherosclerosis/drug therapy , Cell Line, Tumor , Deoxyadenosines/administration & dosage , Deoxyadenosines/antagonists & inhibitors , Deoxyadenosines/pharmacology , Drug Therapy, Combination , Humans , Kupffer Cells/drug effects , Medicine, Chinese Traditional , Methotrexate/therapeutic use , Models, Biological , Pentostatin/administration & dosage , Plant Extracts/chemistry , Plant Extracts/therapeutic use , Receptor, Adenosine A3 , Signal Transduction/drug effects
13.
Meat Sci ; 102: 8-14, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25498483

ABSTRACT

Postmortem energy metabolism drives hydrogen accumulation in muscle and results in a fairly constant ultimate pH. Extended glycolysis results in adverse pork quality and may be possible with greater adenonucleotide availability postmortem. We hypothesized that slowing adenonucleotide removal by reducing AMP deaminase activity would extend glycolysis and lower the ultimate pH of muscle. Longissimus muscle samples were incorporated into an in vitro system that mimics postmortem glycolysis with or without pentostatin, an AMP deaminase inhibitor. Pentostatin lowered ultimate pH and increased lactate and glucose 6-phosphate with time. Based on these results and that AMPK γ3(R200Q) mutated pigs (RN⁻) produce low ultimate pH pork, we hypothesized AMP deaminase abundance and activity would be lower in RN⁻ muscle than wild-type. RN⁻ muscle contained lower AMP deaminase abundance and activity. These data show that altering adenonucleotide availability postmortem can extend postmortem pH decline and suggest that AMP deaminase activity may, in part, contribute to the low ultimate pH observed in RN⁻ pork.


Subject(s)
AMP Deaminase/metabolism , Food Quality , Food Storage , Glycolysis , Meat/analysis , Muscle, Skeletal/enzymology , AMP Deaminase/antagonists & inhibitors , AMP Deaminase/genetics , Adenosine Deaminase Inhibitors/pharmacology , Amino Acid Substitution , Animals , Animals, Inbred Strains , Glycolysis/drug effects , Hydrogen-Ion Concentration , Muscle, Skeletal/chemistry , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Mutation , Pentostatin/pharmacology , Polymorphism, Single Nucleotide , Protein Subunits , Sus scrofa , Virginia
14.
Bone Marrow Transplant ; 49(9): 1198-204, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25046213

ABSTRACT

Extracorporeal photopheresis (ECP) and the purine analog pentostatin exert potent immunomodulatory effects. We evaluated the use of these treatment modalities to prevent GVHD in a canine model of unrelated dog leukocyte Ag-mismatched hematopoietic cell transplantation, after conditioning with 920 cGy TBI. We have shown previously in this model that 36/40 dogs given MTX alone as postgrafting immunosuppression engrafted and that 25 of 40 dogs had severe GVHD and median survival of 21 days. In the current study, nine dogs received conditioning with 920 cGy TBI and postgrafting MTX either with ECP on days -2 to -1 alone (n=5) or ECP on days -6 and -5 combined with two doses of pentostatin (days -4 to -3) (n=4). Seven of nine dogs achieved engraftment. Six dogs developed severe acute GVHD (four in the group with ECP alone and two with pentostatin and ECP). We failed to demonstrate a positive impact of ECP and pentostatin for the prevention of GVHD compared with historical control dogs.


Subject(s)
Antineoplastic Agents/pharmacology , Dog Diseases/therapy , Graft vs Host Disease/veterinary , Hematopoietic Stem Cell Transplantation/methods , Pentostatin/pharmacology , Photopheresis/veterinary , Transplantation Conditioning/methods , Animals , Dog Diseases/prevention & control , Dogs , Flow Cytometry , Graft vs Host Disease/prevention & control , Photopheresis/methods , Transplantation Chimera
15.
Exp Parasitol ; 135(2): 357-62, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23933282

ABSTRACT

This study aimed to verify the effect of 3'-deoxyadenosine and deoxycoformycin on hematologic parameters and adenosine deaminase (ADA) activity in plasma and brain of mice infected with Trypanosoma evansi. Seventy animals were divided into seven groups, which were divided into two subgroups each for sampling on days 4 and 8 post-infection (PI). The groups were composed of three uninfected groups (A-C), namely, not-treated (A), treated with 3'-deoxyadenosine (B), and treated with deoxycoformycin (C) and four infected groups, mice with T. evansi (D-G), namely, not-treated (D), treated with 3'-deoxyadenosine (E), treated with deoxycoformycin (F), and treated with a combination 3'-deoxyadenosine and deoxycoformycin (G). Hematological parameters and ADA activity were evaluated in plasma and brain. Animals in groups B and C exhibited a reduction in the levels of plasma total protein compared group A. Animals in groups D and F showed changes in the hematological parameters. The ADA activity significantly reduced in the animals of groups C, D, F and G. Mice in the group E presented increased ADA activity in plasma. Therefore, we conclude that the treatment interferes significantly in the hematologic parameters in mice infected with T. evansi. On the other hand, when the ADA inhibitor was used we observed a significant decrease in the values of hematocrit, total erythrocytes, and hemoglobin concentration. The deoxycoformycin was able to inhibit the ADA activity of parasite thus it may be one of the mechanisms of efficacy of this treatment.


Subject(s)
Adenosine Deaminase Inhibitors/therapeutic use , Adenosine Deaminase/metabolism , Brain/enzymology , Pentostatin/therapeutic use , Trypanosomiasis/drug therapy , Adenosine Deaminase/blood , Adenosine Deaminase Inhibitors/pharmacology , Animals , Blood Proteins/drug effects , Blood Proteins/metabolism , Brain/drug effects , Deoxyadenosines/antagonists & inhibitors , Deoxyadenosines/pharmacology , Deoxyadenosines/therapeutic use , Dose-Response Relationship, Drug , Erythrocyte Count , Female , Hematocrit , Hemoglobins/analysis , Leukocyte Count , Mice , Parasitemia/drug therapy , Pentostatin/pharmacology , Trypanosoma/drug effects , Trypanosoma/enzymology , Trypanosomiasis/blood , Trypanosomiasis/enzymology
16.
Am J Physiol Lung Cell Mol Physiol ; 304(5): L361-70, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23316066

ABSTRACT

Pulmonary endothelial cell (EC) apoptosis has been implicated in the pathogenesis of emphysema. Cigarette smoke (CS) causes lung EC apoptosis and emphysema. In this study, we show that CS exposure increased lung tissue adenosine levels in mice, an effect associated with increased lung EC apoptosis and the development of emphysema. Adenosine has a protective effect against apoptosis via adenosine receptor-mediated signaling. However, sustained elevated adenosine increases alveolar cell apoptosis in adenosine deaminase-deficient mice. We established an in vitro model of sustained adenosine exposure by incubating lung EC with adenosine in the presence of an adenosine deaminase inhibitor, deoxycoformicin. We demonstrated that sustained adenosine exposure caused lung EC apoptosis via nucleoside transporter-facilitated intracellular adenosine uptake, subsequent activation of p38 and JNK in mitochondria, and ultimately mitochondrial defects and activation of the mitochondria-mediated intrinsic pathway of apoptosis. Our results suggest that sustained elevated adenosine may contribute to CS-induced lung EC apoptosis and emphysema. Our data also reconcile the paradoxical effects of adenosine on apoptosis, demonstrating that prolonged exposure causes apoptosis via nucleoside transporter-mediated intracellular adenosine signaling, whereas acute exposure protects against apoptosis via activation of adenosine receptors. Inhibition of adenosine uptake may become a new therapeutic target in treatment of CS-induced lung diseases.


Subject(s)
Adenosine/metabolism , Apoptosis/drug effects , Endothelial Cells/physiology , Smoke/adverse effects , Adenosine Deaminase/deficiency , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Adenosine Deaminase Inhibitors/pharmacology , Animals , Cattle , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelium/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Lung Injury , Mice , Mice, Inbred AKR , Mice, Inbred C57BL , Mitochondria/metabolism , Nucleoside Transport Proteins/metabolism , Pentostatin/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism
17.
Biol Blood Marrow Transplant ; 19(3): 481-5, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23247045

ABSTRACT

Although significant strides have been made in understanding the biology of graft-versus-host disease (GVHD) and its prevention over the last 4 decades, little is known about the different populations of lymphocytes and the changes in response to treatment for this condition. BMT-CTN 0302 was a randomized phase II clinical trial in the Blood and Marrow Transplant Clinical Trials Network that assessed the efficacy of combination therapy with steroids plus pentostatin, mycophenolate mofetil, etanercept, or denileukin diftitox in patients with acute GVHD. Patients enrolled in the study underwent blood analysis by flow cytometry on days 0, 14, and 28 of therapy to enumerate the number of total lymphocytes, T cells, B cells, and lymphocytes expressing activation markers. Baseline total lymphocyte counts and subpopulations were similar in the 4 treatment arms. Responding patients had a smaller decrease in total CD45(+) cell count (P = .005) compared with nonresponding patients at day 28. On univariate analysis, those who developed chronic GVHD had significantly higher CD8(+) cell counts at day 14 compared with those without it (P = .005). There was no significant association between baseline lymphocyte count and survival. On univariate analysis, among the patients with higher lymphocyte counts at days 14 and 28, there was a trend toward better survival at day 180, although this trend did not reach the predetermined threshold for significance. We found no significant differences in lymphocyte total or subpopulation counts among the 4 treatment arms, and no notable influence on outcomes.


Subject(s)
B-Lymphocyte Subsets/immunology , Bone Marrow Transplantation , Graft vs Host Disease/therapy , T-Lymphocyte Subsets/immunology , Acute Disease , Adult , Antigens, CD/immunology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , B-Lymphocyte Subsets/pathology , Diphtheria Toxin/pharmacology , Diphtheria Toxin/therapeutic use , Etanercept , Female , Graft vs Host Disease/immunology , Graft vs Host Disease/mortality , Graft vs Host Disease/pathology , Humans , Immunoglobulin G/pharmacology , Immunoglobulin G/therapeutic use , Immunophenotyping , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Interleukin-2/pharmacology , Interleukin-2/therapeutic use , Lymphocyte Count , Male , Middle Aged , Mycophenolic Acid/analogs & derivatives , Mycophenolic Acid/pharmacology , Mycophenolic Acid/therapeutic use , Pentostatin/pharmacology , Pentostatin/therapeutic use , Receptors, Tumor Necrosis Factor/therapeutic use , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use , Survival Analysis , T-Lymphocyte Subsets/pathology , Transplantation, Homologous
18.
Am J Respir Cell Mol Biol ; 47(5): 604-13, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22744860

ABSTRACT

Previous studies by our group as well as others have shown that acute adenosine exposure enhances lung vascular endothelial barrier integrity and protects against increased permeability lung edema. In contrast, there is growing evidence that sustained adenosine exposure has detrimental effects on the lungs, including lung edema. It is well established that adenosine modulates lung inflammation. However, little is known concerning the effect of sustained adenosine exposure on lung endothelial cells (ECs), which are critical to the maintenance of the alveolar-capillary barrier. We show that exogenous adenosine plus adenosine deaminase inhibitor caused sustained elevation of adenosine in lung ECs. This sustained adenosine exposure decreased EC barrier function, elevated cellular reactive oxygen species levels, and activated p38, JNK, and RhoA. Inhibition of equilibrative nucleoside transporters (ENTs) prevented sustained adenosine-induced p38 and JNK activation and EC barrier dysfunction. Inhibition of p38, JNK, or RhoA also partially attenuated sustained adenosine-induced EC barrier dysfunction. These data indicate that sustained adenosine exposure causes lung EC barrier dysfunction via ENT-dependent intracellular adenosine uptake and subsequent activation of p38, JNK, and RhoA. The antioxidant N-acetylcysteine and the NADPH inhibitor partially blunted sustained adenosine-induced JNK activation but were ineffective in attenuation of p38 activation or barrier dysfunction. p38 was activated exclusively in mitochondria, whereas JNK was activated in mitochondria and cytoplasm by sustained adenosine exposure. Our data further suggest that sustained adenosine exposure may cause mitochondrial oxidative stress, leading to activation of p38, JNK, and RhoA in mitochondria and resulting in EC barrier dysfunction.


Subject(s)
Adenosine/physiology , Capillary Permeability , Endothelial Cells/metabolism , Equilibrative Nucleoside Transport Proteins/metabolism , Pulmonary Artery/pathology , Signal Transduction , Adenosine/pharmacology , Adenosine Deaminase/metabolism , Adenosine Deaminase Inhibitors/pharmacology , Adherens Junctions/metabolism , Animals , Apoptosis , Cattle , Cells, Cultured , Electric Impedance , Endothelial Cells/drug effects , Endothelial Cells/physiology , Enzyme Activation , JNK Mitogen-Activated Protein Kinases/metabolism , Mitochondria, Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/physiology , Oxidative Stress , Pentostatin/pharmacology , Rats , Reactive Oxygen Species/metabolism , Stress Fibers/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
20.
Leuk Lymphoma ; 52 Suppl 2: 43-5, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21463108

ABSTRACT

Pentostatin (2'-deoxycoformycin; dCF) and cladribine (2-chlorodeoxyadenosine; CdA) are highly effective agents for the treatment of hairy cell leukemia. Although their precise mechanisms of action in this disease are still unknown, a number of mechanisms have been postulated. dCF is a potent inhibitor of adenosine deaminase (ADA), and treatment results in the accumulation of deoxyadenosine (dAdo) and adenosine (Ado) in the plasma. dAdo is phosphorylated by deoxycytidine kinase in lymphocytes to deoxyadenosine monophosphate (dAMP), which is subsequently converted to deoxyadenosine triphosphate (dATP). CdA is the chlorinated derivative of deoxyadenosine, is resistant to degradation by ADA, and accumulates in lymphocytes as CdATP. Both dATP and CdATP cause an initial accumulation of DNA strand breaks in lymphocytes and this results in the activation of p53, the release of cytochrome c from mitochondria, and apoptosis. CdA has several unique mechanisms of action over dAdo and these include the incorportation of CdATP into DNA, the inhibition of DNA polymerase ß, and the phosphorylation of CdA to CdATP by deoxyguanosine kinase in mitochondria. These additional modes of action produce further DNA breaks in CdA-treated cells and explain the more potent activity of CdA compared to dCF and the greater myelosuppression with this agent. The cells die by apoptosis, but the DNA strand breaks also cause the activation of poly(ADP-ribose) polymerase (PARP), with resultant cellular depletion of nicotinamide adenine dinucleotide (NAD) and ATP. The induction of necrosis by PARP activation may explain the activity of these analogs in some patients with p53 mutations.


Subject(s)
Cladribine/pharmacology , Leukemia, Hairy Cell/drug therapy , Pentostatin/pharmacology , Apoptosis/drug effects , Cladribine/therapeutic use , DNA Breaks/drug effects , Humans , Pentostatin/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...