Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
1.
Antimicrob Resist Infect Control ; 11(1): 5, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35012641

ABSTRACT

BACKGROUND: Periprosthetic joint infection (PJI) causes significant morbidity. Methicillin sensitive Staphylococcus aureus (MSSA) is the most frequent organism, and the majority are endogenous. Decolonisation reduces PJIs but there is a paucity of evidence comparing treatments. Aims; compare 3 nasal decolonisation treatments at (1) achieving MSSA decolonisation, (2) preventing PJI. METHODS: Our hospital prospectively collected data on our MSSA decolonisation programme since 2013, including; all MSSA carriers, treatment received, MSSA status at time of surgery and all PJIs. Prior to 2017 MSSA carriers received nasal mupirocin or neomycin, from August 2017 until August 2019 nasal octenidine was used. RESULTS: During the study period 15,958 primary hip and knee replacements were performed. 3200 (20.1%) were MSSA positive at preoperative screening and received decolonisation treatment, 698 mupirocin, 1210 neomycin and 1221 octenidine. Mupirocin (89.1%) and neomycin (90.9%) were more effective at decolonisation than octenidine (50.0%, P < 0.0001). There was no difference in PJI rates (P = 0.452). CONCLUSIONS: Mupirocin and neomycin are more effective than octenidine at MSSA decolonisation. There was poor correlation between the MSSA status after treatment (on day of surgery) and PJI rates. Further research is needed to compare alternative MSSA decolonisation treatments.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Imines/therapeutic use , Mupirocin/therapeutic use , Neomycin/therapeutic use , Nose Diseases/prevention & control , Pyridines/therapeutic use , Staphylococcal Infections/prevention & control , Anti-Infective Agents, Local/therapeutic use , Cohort Studies , Drug Resistance, Bacterial , England , Joint Diseases/microbiology , Joint Diseases/prevention & control , Methicillin/pharmacology , Nose Diseases/microbiology , Retrospective Studies , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology
2.
Biomolecules ; 11(8)2021 08 04.
Article in English | MEDLINE | ID: mdl-34439823

ABSTRACT

Alveolar osteitis (AO) is a common complication following the extraction of the teeth, particularly the lower third molars. It starts within a few days after the extraction and manifests mainly as pain in the extraction site. Several strategies of treatment are available in order to relieve pain and heal the extraction wound. Recently, a novel medical device combining hyaluronic acid (HA) and octenidine (OCT) was introduced for the treatment of AO. This series of case reports aims to summarize the initial clinical experiences with this new device and to highlight factors possibly interfering with this treatment. The medical documentation of five patients with similar initial situations treated for AO with HA + OCT device was analyzed in detail. Smoking and previous treatment with Alveogyl (Septodont, Saint-Maur-des-Fossés, France) were identified as factors interfering with the AO treatment with the HA + OCT device. In three patients without these risk factors, the treatment led to recovery within two or three days. The patient pretreated with Alveogyl and the smoker required six and seven applications of the HA + OCT device, respectively. According to these initial observations, it seems smoking and previous treatment with Alveogyl prolong the treatment of AO using the HA + OCT device that, in turn, shows a rapid effect if these risk factors are not present.


Subject(s)
Dry Socket/drug therapy , Hyaluronic Acid/therapeutic use , Imines/therapeutic use , Pain/drug therapy , Pyridines/therapeutic use , Wound Healing/drug effects , Adolescent , Adult , Creosote/adverse effects , Drug Combinations , Dry Socket/etiology , Dry Socket/physiopathology , Dry Socket/surgery , Equipment and Supplies , Female , Humans , Hydrocarbons, Iodinated/adverse effects , Middle Aged , Molar/surgery , Pain/etiology , Pain/physiopathology , Pain/surgery , Risk Factors , Smoking/adverse effects , Thymol , Tooth Extraction/adverse effects , Treatment Outcome , Wound Healing/physiology
3.
Int J Mol Sci ; 22(8)2021 Apr 16.
Article in English | MEDLINE | ID: mdl-33923802

ABSTRACT

Thromboembolic complications are a leading cause of morbidity and mortality in cancer patients. Cancer patients often present with an increased risk for thrombosis including hypercoagulation, so the application of antiplatelet strategies to oncology warrants further investigation. This study investigated the effects of anastrozole and antiplatelet therapy (aspirin/clopidogrel cocktail or atopaxar) treatment on the tumour responses of luminal phenotype breast cancer cells and induced hypercoagulation. Ethical clearance was obtained (M150263). Blood was co-cultured with breast cancer cell lines (MCF7 and T47D) pre-treated with anastrozole and/or antiplatelet drugs for 24 h. Hypercoagulation was indicated by thrombin production and platelet activation (morphological and molecular). Gene expression associated with the epithelial-to-mesenchymal transition (EMT) was assessed in breast cancer cells, and secreted cytokines associated with tumour progression were evaluated. Data were analysed with the PAST3 software. Our findings showed that antiplatelet therapies (aspirin/clopidogrel cocktail and atopaxar) combined with anastrozole failed to prevent hypercoagulation and induced evidence of a partial EMT. Differences in tumour responses that modulate tumour aggression were noted between breast cancer cell lines, and this may be an important consideration in the clinical management of subphenotypes of luminal phenotype breast cancer. Further investigation is needed before this treatment modality (combined hormone and antiplatelet therapy) can be considered for managing tumour associated-thromboembolic disorder.


Subject(s)
Anastrozole/adverse effects , Antineoplastic Agents, Hormonal/adverse effects , Blood Coagulation , Breast Neoplasms/drug therapy , Epithelial-Mesenchymal Transition , Platelet Aggregation Inhibitors/adverse effects , Thrombophilia/prevention & control , Adult , Anastrozole/administration & dosage , Anastrozole/therapeutic use , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents, Hormonal/therapeutic use , Aspirin/administration & dosage , Aspirin/adverse effects , Aspirin/therapeutic use , Breast Neoplasms/complications , Cells, Cultured , Clopidogrel/administration & dosage , Clopidogrel/adverse effects , Clopidogrel/therapeutic use , Drug Interactions , Female , Humans , Imines/administration & dosage , Imines/adverse effects , Imines/therapeutic use , MCF-7 Cells , Platelet Aggregation Inhibitors/administration & dosage , Platelet Aggregation Inhibitors/therapeutic use , Pyridines/administration & dosage , Pyridines/adverse effects , Pyridines/therapeutic use , Thrombin/metabolism , Thrombophilia/drug therapy , Thrombophilia/etiology
4.
Antimicrob Agents Chemother ; 65(7): e0151320, 2021 06 17.
Article in English | MEDLINE | ID: mdl-33903112

ABSTRACT

Leishmaniasis is one of the most challenging neglected tropical diseases and remains a global threat to public health. Currently available therapies for leishmaniases present significant drawbacks and are rendered increasingly inefficient due to parasite resistance, making the need for more effective, safer, and less expensive drugs an urgent one. In our efforts to identify novel chemical scaffolds for the development of antileishmanial agents, we have screened in-house antiplasmodial libraries against axenic and intracellular forms of Leishmania infantum, Leishmania amazonensis, and Leishmania major. Several of the screened compounds showed half-maximal inhibitory concentrations (IC50s) against intracellular L. infantum parasites in the submicromolar range (compounds 1h, IC50 = 0.9 µM, and 1n, IC50 = 0.7 µM) and selectivity indexes of 11 and 9.7, respectively. Compounds also displayed activity against L. amazonensis and L. major parasites, albeit in the low micromolar range. Mechanistic studies revealed that compound 1n efficiently inhibits oxygen consumption and significantly decreases the mitochondrial membrane potential in L. infantum axenic amastigotes, suggesting that this chemotype acts, at least in part, by interfering with mitochondrial function. Structure-activity analysis suggests that compound 1n is a promising antileishmanial lead and emphasizes the potential of the quinoline-(1H)-imine chemotype for the future development of new antileishmanial agents.


Subject(s)
Antiprotozoal Agents , Leishmania mexicana , Leishmaniasis , Animals , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/therapeutic use , Imines/therapeutic use , Leishmaniasis/drug therapy , Macrophages , Mice , Mice, Inbred BALB C
6.
Mol Pharmacol ; 98(6): 648-657, 2020 12.
Article in English | MEDLINE | ID: mdl-32978326

ABSTRACT

Protein tyrosine phosphatase (PTP) 4A3 is frequently overexpressed in human solid tumors and hematologic malignancies and is associated with tumor cell invasion, metastasis, and a poor patient prognosis. Several potent, selective, and allosteric small molecule inhibitors of PTP4A3 were recently identified. A lead compound in the series, JMS-053 (7-imino-2-phenylthieno[3,2-c]pyridine-4,6(5H,7H)-dione), has a long plasma half-life (∼ 24 hours) in mice, suggesting possible binding to serum components. We confirmed by isothermal titration calorimetry that JMS-053 binds to human serum albumin. A single JMS-053 binding site was identified by X-ray crystallography in human serum albumin at drug site 3, which is also known as subdomain IB. The binding of JMS-053 to human serum albumin, however, did not markedly alter the overall albumin structure. In the presence of serum albumin, the potency of JMS-053 as an in vitro inhibitor of PTP4A3 and human A2780 ovarian cancer cell growth was reduced. The reversible binding of JMS-053 to serum albumin may serve to increase JMS-053's plasma half-life and thus extend the delivery of the compound to tumors. SIGNIFICANCE STATEMENT: X-ray crystallography revealed that a potent, reversible, first-in-class small molecule inhibitor of the oncogenic phosphatase protein tyrosine phosphatase 4A3 binds to at least one site on human serum albumin, which is likely to extend the compound's plasma half-life and thus assist in drug delivery into tumors.


Subject(s)
Imines/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Protein Tyrosine Phosphatases/antagonists & inhibitors , Pyridines/pharmacology , Serum Albumin, Human/metabolism , Binding Sites , Calorimetry , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Enzyme Assays , Half-Life , Humans , Imines/chemistry , Imines/therapeutic use , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Protein Tyrosine Phosphatases/metabolism , Pyridines/chemistry , Pyridines/therapeutic use , Serum Albumin, Human/ultrastructure
7.
Bioorg Med Chem Lett ; 30(1): 126774, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31685339

ABSTRACT

In continuation of our program to discover new potential antifungal agents, a series of amide and imine derivatives containing a kakuol moiety were synthesized and characterized by the spectroscopic analysis. By using the mycelium growth rate method, the target compounds were evaluated systematically for antifungal activities in vitro against four plant pathogenic fungi, and structure-activity relationships (SAR) were derived. Compounds 7d, 7e, 7h, 7i and 7r showed obvious inhibitory activity against the corresponding tested fungi at 50 µg/mL. Especially, compounds 7e and 7r displayed more potent antifungal activity against B. cinerea than that of thiabendazole (a positive control). Moreover, compound 7e also exhibited good activity against A. alternata with EC50 values of 11.0 µg/mL, and the value was slightly superior to that of thiabendazole (EC50 = 14.9 µg/mL). SAR analysis showed that the ether group was a highly sensitive structural moiety to the activity and the type as well as position of substituents on benzene ring could make some effects on the activity.


Subject(s)
Amides/chemical synthesis , Antifungal Agents/therapeutic use , Benzodioxoles/chemical synthesis , Imines/chemical synthesis , Propiophenones/chemical synthesis , Amides/therapeutic use , Antifungal Agents/pharmacology , Benzodioxoles/therapeutic use , Humans , Imines/therapeutic use , Molecular Structure , Propiophenones/therapeutic use , Structure-Activity Relationship
8.
Oxid Med Cell Longev ; 2019: 7180284, 2019.
Article in English | MEDLINE | ID: mdl-31885813

ABSTRACT

Resveratrol (RSV) is a naturally occurring polyphenol that exhibits pleiotropic health benefits, including anticolitis and colon cancer-protective activity. Recently, we identified the novel imine RSV analog (IRA), 2-methoxyl-3,6-dihydroxyl-IRA 3,4,5,4-tetramethoxystilbene (C33), as a putative activator of nuclear factor erythroid 2-related factor 2 (Nrf2). The present study was designed to evaluate the ability of C33 to activate the Nrf2 signaling pathway and its anticolitis effect in comparison to RSV. The anticolitis action of C33 was assessed in a mouse model of colitis induced by dextran sulfate sodium (DSS). The effect of C33 on the Nrf2 signaling pathway was examined in vitro and in vivo. Compared to RSV, C33 triggered a more dramatic increase in the expression of genes downstream of Nrf2 in LS174T cells as well as in the small intestine and colon of wild-type (WT) mice. Correlated with its superior ability to activate the cytoprotective Nrf2 pathway, C33 was significantly better in ameliorating DSS-induced colitis by improving the inflammation score, as well as downregulating the markers of inflammation in WT mice. Moreover, induction of the NOD-like receptors family pyrin domain containing 3 (NLRP3) inflammasome by colitis was also significantly inhibited by the IRA. Nrf2 knockout completely abolished the effects of C33, indicating that Nrf2 is the important mechanistic target of C33 in vivo. In conclusion, the novel IRA, C33, has stronger anticolitis effects than RSV. Further studies are warranted to evaluate C33 as a potential therapeutic agent for inflammatory bowel disease and cancer chemoprevention.


Subject(s)
Imines/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Intestinal Mucosa/metabolism , NF-E2-Related Factor 2/metabolism , Resveratrol/therapeutic use , Animals , Cell Line , Colitis/chemically induced , Dextran Sulfate , Disease Models, Animal , Down-Regulation , Humans , Imines/chemical synthesis , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Resveratrol/analogs & derivatives , Resveratrol/chemical synthesis , Signal Transduction
9.
Mar Drugs ; 17(9)2019 Aug 24.
Article in English | MEDLINE | ID: mdl-31450557

ABSTRACT

In this study, we aimed to find chemicals from lower sea animals with defensive effects against human immunodeficiency virus type 1 (HIV-1). A library of marine natural products consisting of 80 compounds was screened for activity against HIV-1 infection using a luciferase-encoding HIV-1 vector. We identified five compounds that decreased luciferase activity in the vector-inoculated cells. In particular, portimine, isolated from the benthic dinoflagellate Vulcanodinium rugosum, exhibited significant anti-HIV-1 activity. Portimine inhibited viral infection with an 50% inhibitory concentration (IC50) value of 4.1 nM and had no cytotoxic effect on the host cells at concentrations less than 200 nM. Portimine also inhibited vesicular stomatitis virus glycoprotein (VSV-G)-pseudotyped HIV-1 vector infection. This result suggested that portimine mainly targeted HIV-1 Gag or Pol protein. To analyse which replication steps portimine affects, luciferase sequences were amplified by semi-quantitative PCR in total DNA. This analysis revealed that portimine inhibits HIV-1 vector infection before or at the reverse transcription step. Portimine has also been shown to have a direct effect on reverse transcriptase using an in vitro reverse transcriptase assay. Portimine efficiently inhibited HIV-1 replication and is a potent lead compound for developing novel therapeutic drugs against HIV-1-induced diseases.


Subject(s)
Anti-HIV Agents/pharmacology , Aquatic Organisms/chemistry , Dinoflagellida/chemistry , HIV Infections/drug therapy , HIV-1/drug effects , Imines/pharmacology , Spiro Compounds/pharmacology , Anti-HIV Agents/isolation & purification , Anti-HIV Agents/therapeutic use , Drug Evaluation, Preclinical , HEK293 Cells , HIV-1/physiology , HeLa Cells , Humans , Imines/isolation & purification , Imines/therapeutic use , Inhibitory Concentration 50 , Spiro Compounds/isolation & purification , Spiro Compounds/therapeutic use , Virus Replication/drug effects
10.
J Med Chem ; 62(4): 2184-2201, 2019 02 28.
Article in English | MEDLINE | ID: mdl-30715875

ABSTRACT

We describe herein the synthesis and neuroprotective capacity of an array of 31 compounds comprising quinolyloximes, quinolylhydrazones, quinolylimines, QNs, and related heterocyclic azolylnitrones. Neuronal cultures subjected to oxygen-glucose deprivation (OGD), as experimental model for ischemic conditions, were treated with our molecules at the onset of recovery period after OGD and showed that most of these QNs, but not the azo molecules, improved neuronal viability 24 h after recovery. Especially, QN ( Z)- N-tert-butyl-1-(2-chloro-6-methoxyquinolin-3-yl)methanimine oxide (23) was shown as a very potent neuroprotective agent. Antioxidant analysis based on the ability of QN 23 to trap different types of toxic radical oxygenated species supported and confirmed its strong neuroprotective capacity. Finally, QN 23 showed also neuroprotection induction in two in vivo models of cerebral ischemia, decreasing neuronal death and reducing infarct size, allowing us to conclude that QN 23 can be considered as new lead-compound for ischemic stroke treatment.


Subject(s)
Free Radical Scavengers/therapeutic use , Imines/therapeutic use , Ischemic Attack, Transient/drug therapy , Neuroprotective Agents/therapeutic use , Quinolines/therapeutic use , Animals , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Apoptosis/drug effects , Cells, Cultured , Free Radical Scavengers/chemical synthesis , Free Radical Scavengers/pharmacology , Imines/chemical synthesis , Imines/pharmacology , Lipid Peroxidation/drug effects , Male , Mice, Inbred C57BL , Neurons/drug effects , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/pharmacology , Quinolines/chemical synthesis , Quinolines/pharmacology , Rats, Wistar , Reactive Oxygen Species/metabolism
12.
Biotechnol Prog ; 34(1): 249-261, 2018 01.
Article in English | MEDLINE | ID: mdl-28699244

ABSTRACT

Targeted delivery of drug molecules to specific cells in mammalian systems demonstrates a great potential to enhance the efficacy of current pharmaceutical therapies. Conventional strategies for pharmaceutical delivery are often associated with poor therapeutic indices and high systemic cytotoxicity, and this result in poor disease suppression, low surviving rates, and potential contraindication of drug formulation. The emergence of aptamers has elicited new research interests into enhanced targeted drug delivery due to their unique characteristics as targeting elements. Aptamers can be engineered to bind to their cognate cellular targets with high affinity and specificity, and this is important to navigate active drug molecules and deliver sufficient dosage to targeted malignant cells. However, the targeting performance of aptamers can be impacted by several factors including endonuclease-mediated degradation, rapid renal filtration, biochemical complexation, and cell membrane electrostatic repulsion. This has subsequently led to the development of smart aptamer-immobilized biopolymer systems as delivery vehicles for controlled and sustained drug release to specific cells at effective therapeutic dosage and minimal systemic cytotoxicity. This article reports the synthesis and in vitro characterization of a novel multi-layer co-polymeric targeted drug delivery system based on drug-loaded PLGA-Aptamer-PEI (DPAP) formulation with a stage-wise delivery mechanism. A thrombin-specific DNA aptamer was used to develop the DPAP system while Bovine Serum Albumin (BSA) was used as a biopharmaceutical drug in the synthesis process by ultrasonication. Biophysical characterization of the DPAP system showed a spherical shaped particulate formulation with a unimodal particle size distribution of average size ∼0.685 µm and a zeta potential of +0.82 mV. The DPAP formulation showed a high encapsulation efficiency of 89.4 ± 3.6%, a loading capacity of 17.89 ± 0.72 mg BSA protein/100 mg PLGA polymeric particles, low cytotoxicity and a controlled drug release characteristics in 43 days. The results demonstrate a great promise in the development of DPAP formulation for enhanced in vivo cell targeting. © 2017 American Institute of Chemical Engineers Biotechnol. Prog., 34:249-261, 2018.


Subject(s)
Aptamers, Nucleotide/chemistry , Biophysical Phenomena , Drug Delivery Systems , Nanoparticles/chemistry , Animals , Aptamers, Nucleotide/therapeutic use , Cell Movement/drug effects , Humans , Imines/chemistry , Imines/therapeutic use , Nanoparticles/therapeutic use , Particle Size , Polyethylenes/chemistry , Polyethylenes/therapeutic use , Serum Albumin, Bovine/chemistry
13.
Biochimie ; 138: 70-81, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28454919

ABSTRACT

In spite of the tremendous stride in modern medicine, conventional drugs used in the hepatotoxic management are mostly inadequate. The present study aims in the synthesis of novel Schiff base compound derived using s-allyl cystiene and methionine. The newly synthesized compound, 2-((2-((2-(allylthio)-1-carboxyethyl)imino)ethylidene)amino)-4-(methylthio)butanoic acid (ACEMB) was characterized using UV-visible spectrophotometer, FTIR, 1HNMR, and GC-MS. ACEMB showed potent in vitro antioxidant property. Chronic administration of ACEMB prior to CCl4 intoxication: i) attenuated the leakage of liver injury markers, such as, enzymes (AST, ALT, GGT, ALP and LDH) and biomolecules (bilirubin) into the blood circulation; ii) normalized the concentration of total proteins, albumin and globulin to control level; and iii) protected the liver against dyslipidemia. These effects of ACEMB show the preservation of endoplasmic reticulum function against CCl4 toxicity in the liver. The protective effect of ACEMB was due to its antioxidant property, which was revealed by reduced oxidative stress (TBARS and HP) and enhanced functions of the endogenous antioxidative system (SOD, catalase, GPx, GST, GSH, vitamin E and C) against CCl4 intoxication. Also, ACEMB protected the functional activities of the various mitochondrial tricarboxylic acid cycle and oxidative phosphorylation enzymes. The biochemical alterations are in concurrence with the histological observations, wherein ACEMB pretreatment prevented the vacuolation, degeneration of nuclei and necrosis of hepatocytes. In addition, in silico analysis reveals the interaction of ACEMB in the active site of cytochrome P450. ACEMB mediates hepatoprotective effect by substituting itself as an antioxidant and decreasing oxidative stress, thereby diminishing the intracellular organelle dysfunction against CCl4 toxicity in the liver.


Subject(s)
Antioxidants/therapeutic use , Carbon Tetrachloride Poisoning/complications , Cysteine/analogs & derivatives , Imines/therapeutic use , Liver Diseases/drug therapy , Animals , Antioxidants/chemical synthesis , Binding Sites , Catalytic Domain , Cysteine/chemical synthesis , Cysteine/therapeutic use , Cytochrome P-450 CYP2E1/metabolism , Cytochrome P-450 CYP3A/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Imines/chemical synthesis , Liver Diseases/etiology , Liver Diseases/metabolism , Male , Molecular Docking Simulation , Oxidative Stress/drug effects , Rats
14.
Am J Ther ; 24(6): e730-e736, 2017.
Article in English | MEDLINE | ID: mdl-26398717

ABSTRACT

Protease-activated receptor (PAR)-1 inhibitors have recently become popular in the use of atherosclerosis among clinicians. Atherosclerosis can cause cardiovascular and cerebrovascular events leading to one of the major causes of mortality worldwide. Thrombin-mediated platelets can cause atherosclerotic plaques, and these platelets are activated by thrombin through the PAR-1. Vorapaxar and atopaxar are novel antiplatelet drugs that inhibit the thrombin-induced platelet activation by antagonizing the PAR-1. The objective of this article is to review the mechanism of action of vorapaxar and atopaxar and explain the rationale for using them in atherothrombosis patients including myocardial infarction, peripheral arterial disease, and stroke.


Subject(s)
Atherosclerosis/drug therapy , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Receptor, PAR-1/antagonists & inhibitors , Thrombosis/prevention & control , Atherosclerosis/complications , Blood Platelets/drug effects , Blood Platelets/metabolism , Clinical Trials as Topic , Humans , Imines/pharmacology , Imines/therapeutic use , Lactones/pharmacology , Lactones/therapeutic use , Myocardial Infarction/etiology , Myocardial Infarction/prevention & control , Peripheral Arterial Disease/etiology , Peripheral Arterial Disease/prevention & control , Platelet Aggregation Inhibitors/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Receptor, PAR-1/metabolism , Stroke/etiology , Stroke/prevention & control , Thrombin/metabolism , Thrombosis/etiology , Treatment Outcome
15.
Behav Brain Res ; 319: 188-199, 2017 02 15.
Article in English | MEDLINE | ID: mdl-27888018

ABSTRACT

Nitric oxide (NO) is involved in neuronal modifications, and overproduction of NO contributes to memory deficits after acute hypobaric hypoxia-reoxygenation. This study investigated the ability of the iNOS inhibitor 1400W to counteract spatial memory deficits following acute hypobaric hypoxia-reoxygenation, and to affect expression of NOS, NO, 3-NT and MDA production, and apoptosis in rat cerebral cortex. We also used primary rat microglia to investigate the effect of 1400W on expression of NOS, NO, 3-NT and MDA production, and apoptosis. Acute hypobaric hypoxia-reoxygenation impaired spatial memory, and was accompanied by activated microglia, increased iNOS expression, NO, 3-NT and MDA production, and neuronal cell apoptosis in rat cerebral cortex one day post-reoxygenation. 1400W treatment inhibited iNOS expression without affecting nNOS or eNOS. 1400W also reduced NO, 3-NT and MDA production, and prevented neuronal cell apoptosis in cerebral cortex, in addition to reversing spatial memory impairment after acute hypobaric hypoxia-reoxygenation. Hypoxia-reoxygenation activated primary microglia, and increased iNOS and nNOS expression, NO, 3-NT, and MDA production, and apoptosis. Treatment with 1400W inhibited iNOS expression without affecting nNOS, reduced NO, 3-NT and MDA production, and prevented apoptosis in primary microglia. Based on the above findings, we concluded that the highly selective iNOS inhibitor 1400W inhibited iNOS induction in microglial cells, and reduced generation of NO, thereby mitigating oxidative stress and neuronal cell apoptosis in the rat cerebral cortex, and improving the spatial memory dysfunction caused by acute hypobaric hypoxia-reoxygenation.


Subject(s)
Cognition Disorders , Gene Expression Regulation/drug effects , Imines/pharmacology , Imines/therapeutic use , Microglia/enzymology , Nitric Oxide Synthase Type II/metabolism , Animals , Annexin A5/metabolism , Apoptosis/drug effects , Cells, Cultured , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Cognition Disorders/pathology , Disease Models, Animal , Gene Expression Regulation/physiology , Hypoxia/complications , Lipid Peroxidation/drug effects , Malondialdehyde/metabolism , Maze Learning/drug effects , Nerve Tissue Proteins/metabolism , Nitric Oxide/metabolism , Oxygen/pharmacology , Rats , Rats, Sprague-Dawley , Tyrosine/analogs & derivatives , Tyrosine/metabolism
16.
Curr Pharm Des ; 22(29): 4610-4616, 2016.
Article in English | MEDLINE | ID: mdl-27281329

ABSTRACT

BACKGROUND: Acetylsalicylic acid and clopidogrel are two antiplatelet agents currently used in the therapy of peripheral arterial disease. Cilostazol also inhibits platelet aggegration. These agents present limitations that novel antiplatelet agents may overcome. OBJECTIVE: The aim of this manuscript is to review current data on the use of novel antiplatelet agents in peripheral arterial disease. METHOD: An extensive search in the English medical literature has yielded a number of publications on a number of novel antiplatelet agents; atopaxar, vorapaxar, cangrelor, ticagrelor, elinogrel, and prasugrel. RESULTS: Data on atopaxar, vorapaxar, cangrelor, ticagrelor, elinogrel and prasugrel come mainly from cardiology publications. Limitations, side effects and effectiveness of each of these agents are studied, but their use in peripheral arterial disease is limited, especially for those agents that have not still been approved for this indication. As expected, main side effect of most of these agents is haemorrhage, but other important side effects limit the use of some of these agents in specific subgroups of patients. CONCLUSION: Novel antiplatelet agents demonstrate a range of promising characteristics, but further study and clinical trials are necessary for them to be considered safe and effective.


Subject(s)
Peripheral Arterial Disease/drug therapy , Platelet Aggregation Inhibitors/therapeutic use , Adenosine/analogs & derivatives , Adenosine/therapeutic use , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/therapeutic use , Imines/therapeutic use , Lactones/therapeutic use , Prasugrel Hydrochloride/therapeutic use , Pyridines/therapeutic use , Quinazolinones/therapeutic use , Sulfonamides/therapeutic use , Ticagrelor
17.
ChemMedChem ; 11(12): 1318-27, 2016 Jun 20.
Article in English | MEDLINE | ID: mdl-26804623

ABSTRACT

Given the complex nature of Alzheimer's disease (AD), compounds that are able to simultaneously address two or more AD-associated targets show greater promise for development into drugs for AD therapy. Herein we report an efficient two-step synthesis and biological evaluation of new racemic benzochromene derivatives as antioxidants, inhibitors of cholinesterase and ß-amyloid (Aß1-42 ) aggregation. Based on the results of the primary screening, we identified 15-(3-methoxyphenyl)-9,11,12,15-tetrahydro-10H,14H-benzo[5,6]chromeno[2,3-d]pyrido[1,2-a]pyrimidin-14-imine (3 e) and 16-(3-methoxyphenyl)-9,10,11,12,13,16-hexahydro-15H-benzo[5',6']chromeno[2',3':4,5]pyrimido[1,2-a]azepin-15-imine (3 f) as new potential multitarget-directed ligands for AD therapy. Further in-depth biological analysis showed that compound 3 f is a good human acetylcholinesterase inhibitor [IC50 =(0.36±0.02) µm], has strong antioxidant activity (3.61 µmol Trolox equivalents), and moderate Aß1-42 antiaggregating power (40.3 %).


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Antioxidants/chemical synthesis , Cholinesterase Inhibitors/therapeutic use , Cholinesterases/metabolism , Imines/chemistry , Peptide Fragments/metabolism , Amyloid beta-Peptides/antagonists & inhibitors , Antioxidants/chemistry , Antioxidants/therapeutic use , Binding Sites , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Cholinesterases/chemistry , Humans , Imines/chemical synthesis , Imines/therapeutic use , Ligands , Molecular Docking Simulation , Peptide Fragments/antagonists & inhibitors , Protein Structure, Tertiary
18.
PLoS One ; 10(8): e0134276, 2015.
Article in English | MEDLINE | ID: mdl-26248280

ABSTRACT

Pediatric glioblastomas (pGBM), although rare, are one of the leading causes of cancer-related deaths in children, with tumors essentially refractory to existing treatments. Here, we describe the use of conventional and advanced in vivo magnetic resonance imaging (MRI) techniques to assess a novel orthotopic xenograft pGBM mouse (IC-3752GBM patient-derived culture) model, and to monitor the effects of the anti-cancer agent OKN-007 as an inhibitor of pGBM tumor growth. Immunohistochemistry support data is also presented for cell proliferation and tumor growth signaling. OKN-007 was found to significantly decrease tumor volumes (p<0.05) and increase animal survival (p<0.05) in all OKN-007-treated mice compared to untreated animals. In a responsive cohort of treated animals, OKN-007 was able to significantly decrease tumor volumes (p<0.0001), increase survival (p<0.001), and increase diffusion (p<0.01) and perfusion rates (p<0.05). OKN-007 also significantly reduced lipid tumor metabolism in responsive animals [(Lip1.3 and Lip0.9)-to-creatine ratio (p<0.05)], as well as significantly decrease tumor cell proliferation (p<0.05) and microvessel density (p<0.05). Furthermore, in relationship to the PDGFRα pathway, OKN-007 was able to significantly decrease SULF2 (p<0.05) and PDGFR-α (platelet-derived growth factor receptor-α) (p<0.05) immunoexpression, and significantly increase decorin expression (p<0.05) in responsive mice. This study indicates that OKN-007 may be an effective anti-cancer agent for some patients with pGBMs by inhibiting cell proliferation and angiogenesis, possibly via the PDGFRα pathway, and could be considered as an additional therapy for pediatric brain tumor patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzenesulfonates/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Imines/therapeutic use , Animals , Antineoplastic Agents/toxicity , Benzenesulfonates/toxicity , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Child , Decorin/metabolism , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Imines/toxicity , Immunohistochemistry , Mice , Mice, Nude , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction/drug effects , Sulfatases/metabolism , Survival Rate , Transplantation, Heterologous , Tumor Cells, Cultured
19.
Exp Biol Med (Maywood) ; 240(4): 498-507, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25519430

ABSTRACT

A feature of allergic airway disease is the observed increase of nitric oxide (NO) in exhaled breath. Gram-negative bacterial infections have also been linked with asthma exacerbations. However, the role of NO in asthma exacerbations with gram-negative bacterial infections is still unclear. In this study, we examined the role of NO in lipopolysaccharide (LPS)-induced inflammation in an ovalbumin (OVA)-challenged mouse asthma model. To determine whether NO affected the LPS-induced response, a NO donor (S-nitroso-N-acetylpenicillamine, SNAP) or a selective inhibitor of NO synthase (1400W) was injected intraperitoneally into the mice before the LPS stimulation. Decreased levels of proinflammatory cytokines were demonstrated in the bronchoalveolar lavage fluid from mice treated with SNAP, whereas increased levels of cytokines were found in the 1400W-treated mice. To further explore the molecular mechanism of NO-mediated inhibition of proinflammatory responses in macrophages, RAW 264.7 cells were treated with 1400W or SNAP before LPS stimulation. LPS-induced inflammation in the cells was attenuated by the presence of NO. The LPS-induced IκB kinase (IKK) activation and the expression of IKK were reduced by NO through attenuation of the interaction between Hsp90 and IKK in the cells. The IKK decrease in the lung immunohistopathology was verified in SNAP-treated asthma mice, whereas IKK increased in the 1400W-treated group. We report for the first time that NO attenuates the interaction between Hsp90 and IKK, decreasing the stability of IKK and causing the down-regulation of the proinflammatory response. Furthermore, the results suggest that NO may repress LPS-stimulated innate immunity to promote pulmonary bacterial infection in asthma patients.


Subject(s)
Asthma/prevention & control , HSP90 Heat-Shock Proteins/physiology , I-kappa B Kinase/physiology , Inflammation/prevention & control , Nitric Oxide/pharmacology , Nitric Oxide/therapeutic use , Signal Transduction/physiology , Animals , Asthma/chemically induced , Asthma/physiopathology , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Female , Imines/pharmacology , Imines/therapeutic use , Inflammation/chemically induced , Inflammation/physiopathology , Lipopolysaccharides/adverse effects , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred BALB C , Nitric Oxide Synthase/antagonists & inhibitors , Ovalbumin/adverse effects , S-Nitroso-N-Acetylpenicillamine/pharmacology , S-Nitroso-N-Acetylpenicillamine/therapeutic use , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...