Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.847
Filter
1.
Sci Rep ; 14(1): 15178, 2024 07 10.
Article in English | MEDLINE | ID: mdl-38987553

ABSTRACT

The evolution of endovascular therapies, particularly in the field of intracranial aneurysm treatment, has been truly remarkable and is characterized by the development of various stents. However, ischemic complications related to thrombosis or downstream emboli pose a challenge for the broader clinical application of such stents. Despite advancements in surface modification technologies, an ideal coating that fulfills all the desired requirements, including anti-thrombogenicity and swift endothelialization, has not been available. To address these issues, we investigated a new coating comprising 3-aminopropyltriethoxysilane (APTES) with both anti-thrombogenic and cell-adhesion properties. We assessed the anti-thrombogenic property of the coating using an in vitro blood loop model by evaluating the platelet count and the level of the thrombin-antithrombin (TAT) complex, and investigating thrombus formation on the surface using scanning electron microscopy (SEM). We then assessed endothelial cell adhesion on the metal surfaces. In vitro blood tests revealed that, compared to a bare stent, the coating significantly inhibited platelet reduction and thrombus formation; more human serum albumin spontaneously adhered to the coated surface to block thrombogenic activation in the blood. Cell adhesion tests also indicated a significant increase in the number of cells adhering to the APTES-coated surfaces compared to the numbers adhering to either the bare stent or the stent coated with an anti-fouling phospholipid polymer. Finally, we performed an in vivo safety test by implanting coated stents into the internal thoracic arteries and ascending pharyngeal arteries of minipigs, and subsequently assessing the health status and vessel patency of the arteries by angiography over the course of 1 week. We found that there were no adverse effects on the pigs and the vascular lumens of their vessels were well maintained in the group with APTES-coated stents. Therefore, our new coating exhibited both high anti-thrombogenicity and cell-adhesion properties, which fulfill the requirements of an implantable stent.


Subject(s)
Cell Adhesion , Coated Materials, Biocompatible , Propylamines , Silanes , Stents , Thrombosis , Silanes/chemistry , Silanes/pharmacology , Animals , Cell Adhesion/drug effects , Humans , Stents/adverse effects , Swine , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Propylamines/pharmacology , Propylamines/chemistry , Adsorption , Thrombosis/prevention & control , Fibrinolytic Agents/pharmacology , Fibrinolytic Agents/chemistry , Blood Platelets/drug effects , Blood Platelets/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism
2.
Steroids ; 198: 109272, 2023 10.
Article in English | MEDLINE | ID: mdl-37468115

ABSTRACT

Allopregnanolone (AP) is a neurosteroid synthesized in the brain and a positive allosteric modulator of γ-aminobutyric acid (GABA) type A receptors. Some drugs possessing the aryloxypropanamine (AOPA) pharmacophore, such as fluoxetine, exert their central nervous system (CNS) effects by increasing the brain AP. Although duloxetine (DLX), dapoxetine (DPX), atomoxetine (ATX) and propranolol (PRL) also possess the AOPA pharmacophore and are used to treat some psychiatric disorders, the capabilities of these drugs to increase the brain AP and the possible involvement of AP in their CNS effects remain to be fully elucidated. To clarify these points, we first developed a method for quantifying AP in the rat brain by liquid chromatography/electrospray ionization-tandem mass spectrometry. Analysis of the changes in the brain AP levels using this method revealed that the intraperitoneal administration of DLX (10 mg/kg), DPX (10 mg/kg) and PRL (20 mg/kg) significantly increased the brain AP (DLX: < 0.40-2.74 ng/g tissue, DPX: 1.48-3.83 ng/g tissue and PRL: < 0.40-2.09 ng/g tissue) compared to the saline administration (<0.40 ng/g tissue). These results suggested the possible involvement of the GABAergic neurosteroid, AP, in the central actions of DLX, DPX and PRL. In contrast, ATX (10 mg/kg) did not affect the AP levels in the brain. In addition, the brain and serum AP levels had a remarkably high positive correlation after the administration of DLX, DPX and PRL. Thus, this study proposed the AP-related novel mechanism of actions of DLX, DPX and PRL in the CNS.


Subject(s)
Neurosteroids , Pregnanolone , Animals , Rats , Brain , Duloxetine Hydrochloride/pharmacology , Pharmaceutical Preparations , Pharmacophore , Pregnanolone/pharmacology , Propranolol/pharmacology , Propylamines/chemistry , Propylamines/pharmacology
3.
Pharmacol Rep ; 75(2): 342-357, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36787018

ABSTRACT

BACKGROUND: Methylphenidate and atomoxetine are used for the treatment of attention-deficit/hyperactivity disorder (ADHD). Our previous studies established the validity of the 6-hydroxydopamine (6-OHDA) mouse model of ADHD and demonstrated hypersensitivity to pain, in line with clinical reports in ADHD patients. Acute methylphenidate treatment reduces hyperactivity and increases attention, but does not affect pain behaviors in this mouse model. Whereas atomoxetine has been shown to be effective against some symptoms of ADHD, nothing is known about its possible action on comorbid pain hypersensitivity. The objectives of the present research are (1) to investigate the effects of acute and chronic treatment with atomoxetine on ADHD-like symptoms and nociceptive thresholds, and (2) to explore the catecholaminergic systems underlying these effects. METHODS: Sham and 6-OHDA cohorts of male mice were tested for hyperactivity (open field), attention and impulsivity (5-choice serial reaction time task test), and thermal (hot plate test) and mechanical (von Frey test) thresholds after acute or repeated treatment with vehicle or atomoxetine (1, 3 or 10 mg/kg). RESULTS: Acute administration of atomoxetine (10 mg/kg) reduced the hyperactivity and impulsivity displayed by 6-OHDA mice, without affecting attention or nociception. However, atomoxetine administered at 3 mg/kg/day for 7 days alleviated the ADHD-like core symptoms and attenuated the hyperalgesic responses. Furthermore, hyperlocomotion and anti-hyperalgesic activity were antagonized with phentolamine, propranolol, and sulpiride pre-treatments. CONCLUSION: These findings demonstrated that when administered chronically, atomoxetine has a significant effect on ADHD-associated pain hypersensitization, likely mediated by both α- and ß-adrenergic and D2/D3 dopaminergic receptors, and suggest new indications for atomoxetine that will need to be confirmed by well-designed clinical trials.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Methylphenidate , Male , Mice , Animals , Atomoxetine Hydrochloride/pharmacology , Atomoxetine Hydrochloride/therapeutic use , Attention Deficit Disorder with Hyperactivity/drug therapy , Attention Deficit Disorder with Hyperactivity/chemically induced , Oxidopamine , Propylamines/pharmacology , Pain/drug therapy , Comorbidity , Adrenergic Uptake Inhibitors/adverse effects
4.
Curr Top Behav Neurosci ; 57: 321-362, 2022.
Article in English | MEDLINE | ID: mdl-35606638

ABSTRACT

Based on core symptoms of inattention and deficient impulse control, and the identification of effective pharmacotherapies such as amphetamine (AMP; Adderall®), methylphenidate (MPH; Ritalin®), and atomoxetine (ATX; Strattera®), ADHD is a clinical condition which provides opportunity for translational research. Neuropsychological tests such as the 5-Choice and Continuous Performance Tasks, which measure aspects of attention and impulse control in animals and humans, provide scope for both forward (animal to human) and reverse (human to animal) translation. Rodent studies support pro-attentive effects of AMP and MPH and effectiveness in controlling some forms of impulsive behavior. In contrast, any pro-attentive effects of ATX appear to be less consistent, the most reliable effects of ATX are recorded in tests of impulsivity. These differences may account for AMP and MPH being recognized as first-line treatments for ADHD with a higher efficacy relative to ATX. DSM-5 classifies three "presentations" of ADHD: predominantly inattentive type (ADHD-I), predominantly hyperactive/impulsive type (ADHD-HI), or combined (ADHD-C). Presently, it is unclear whether AMP, MPH, or ATX has differential levels of efficacy across these presentation types. Nonetheless, these studies encourage confidence for the forward translation of NCEs in efforts to identify newer pharmacotherapies for ADHD.


Subject(s)
Attention Deficit Disorder with Hyperactivity , Central Nervous System Stimulants , Methylphenidate , Animals , Humans , Atomoxetine Hydrochloride/pharmacology , Atomoxetine Hydrochloride/therapeutic use , Attention Deficit Disorder with Hyperactivity/drug therapy , Central Nervous System Stimulants/pharmacology , Central Nervous System Stimulants/therapeutic use , Cognition , Methylphenidate/pharmacology , Methylphenidate/therapeutic use , Propylamines/pharmacology , Propylamines/therapeutic use
5.
Biochem Biophys Res Commun ; 586: 157-162, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34847441

ABSTRACT

Sarcopenia is the age-related loss of muscle mass and function and no pharmacological medication has been approved for its treatment. We established an atrogin-1/MAFbx promoter assay to find drug candidates that inhibit myotube atrophy. Alverine citrate (AC) was identified using high-throughput screening of an existing drug library. AC is an established medicine for stomach and intestinal spasms. AC treatment increased myotube diameter and inhibited atrophy signals induced by either C26-conditioned medium or dexamethasone in cultured C2C12 myoblasts. AC also enhanced myoblast fusion through the upregulation of fusion-related genes during C2C12 myoblast differentiation. Oral administration of AC improves muscle mass and physical performance in aged mice, as well as hindlimb-disused mice. Taken together, our data suggest that AC may be a novel therapeutic candidate for improving muscle weakness, including sarcopenia.


Subject(s)
Aging/genetics , Cell Differentiation/drug effects , Muscular Atrophy/prevention & control , Parasympatholytics/pharmacology , Propylamines/pharmacology , Sarcopenia/prevention & control , Aging/metabolism , Animals , Biomarkers/metabolism , Cadherins/genetics , Cadherins/metabolism , Caveolin 3/genetics , Caveolin 3/metabolism , Cell Line , Dexamethasone/pharmacology , Disease Models, Animal , Gene Expression , High-Throughput Screening Assays , Immobilization , Integrin beta1/genetics , Integrin beta1/metabolism , Mice , Mice, Inbred C57BL , Muscle Development/genetics , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle Strength/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Myoblasts/drug effects , Myoblasts/metabolism , Myoblasts/pathology , Sarcopenia/genetics , Sarcopenia/metabolism , Sarcopenia/pathology
6.
Food Funct ; 12(23): 11926-11937, 2021 Nov 29.
Article in English | MEDLINE | ID: mdl-34739536

ABSTRACT

Gastrointestinal motility (GI) disorder causes symptoms such as dyspepsia, abdominal distention, and constipation and severely affects quality of life. The calcium (Ca2+)-sensing receptor (CaSR) expressed in the digestive tract can be activated by amino acids and participates in GI motility regulation. This study is designed to explore the effect and underlying mechanism of CaSR agonist R568 on the small intestine motility of mice in vivo and ex vivo. R568 was given to male C57BL/6 mice by gavage or incubated with isolated jejunum and ileum segments to observe its effects on GI motility and the involved neurons, neurotransmitters and hormones were detected by fluorescence immunohistochemistry and enzyme-linked immunosorbent assays. The in vivo results showed that the intestinal propulsive rate reduced in response to oral intake of R568. R568 treatment increased the numbers of nitric oxide synthase-positive neurons and nitric oxide release but decreased the choline acetyl transferase-positive neurons and acetylcholine release in the myenteric plexuses. R568 increased the secretion of cholecystokinin in the intestinal tissues and serum but had no effect on the secretion of glucagon like peptide-1. Ex vivo results showed that R568 inhibited the contractility of intestinal strips from the jejunum and ileum. Nitric oxide synthase (NOS) inhibitor L-nitroarginine methyl ester (L-NAME), M-receptor antagonist-atropine, and tetrodotoxin (TTX) failed to block the effect of R568. CaSR co-expressed with interstitial cells of Cajal (ICCs) in the myenteric plexus suggests the possibility that ICCs mediated the effect of R568. Our findings demonstrate that CaSR activation inhibited intestinal motility, and both the enteric nervous system and non-neural mechanism are involved in this process.


Subject(s)
Gastrointestinal Motility/drug effects , Jejunum , Phenethylamines/pharmacology , Propylamines/pharmacology , Receptors, Calcium-Sensing , Acetylcholine/metabolism , Animals , Ileum/drug effects , Ileum/metabolism , Jejunum/drug effects , Jejunum/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/metabolism
7.
Sci Rep ; 11(1): 20827, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34675338

ABSTRACT

Non-alcoholic steatohepatitis (NASH) is a rising health challenge, with no approved drugs. We used a computational drug repositioning strategy to uncover a novel therapy for NASH, identifying a GABA-B receptor agonist, AZD3355 (Lesogaberan) previously evaluated as a therapy for esophageal reflux. AZD3355's potential efficacy in NASH was tested in human stellate cells, human precision cut liver slices (hPCLS), and in vivo in a well-validated murine model of NASH. In human stellate cells AZD3355 significantly downregulated profibrotic gene and protein expression. Transcriptomic analysis of these responses identified key regulatory nodes impacted by AZD3355, including Myc, as well as MAP and ERK kinases. In PCLS, AZD3355 down-regulated collagen1α1, αSMA and TNF-α mRNAs as well as secreted collagen1α1. In vivo, the drug significantly improved histology, profibrogenic gene expression, and tumor development, which was comparable to activity of obeticholic acid in a robust mouse model of NASH, but awaits further testing to determine its relative efficacy in patients. These data identify a well-tolerated clinical stage asset as a novel candidate therapy for human NASH through its hepatoprotective, anti-inflammatory and antifibrotic mechanisms of action. The approach validates computational methods to identify novel therapies in NASH in uncovering new pathways of disease development that can be rapidly translated into clinical trials.


Subject(s)
Drug Repositioning , GABA-B Receptor Agonists/therapeutic use , Liver/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Phosphinic Acids/therapeutic use , Propylamines/therapeutic use , Adult , Aged , Animals , Cell Line , Disease Models, Animal , Female , GABA-B Receptor Agonists/pharmacology , Humans , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Phosphinic Acids/pharmacology , Propylamines/pharmacology
8.
FASEB J ; 35(10): e21874, 2021 10.
Article in English | MEDLINE | ID: mdl-34486176

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD), caused by mutations of PKD1 or PKD2 genes, is characterized by development and growth of cysts causing progressive kidney enlargement. Reduced resting cytosolic calcium and increased cAMP levels associated with the tonic action of vasopressin are two central biochemical defects in ADPKD. Here we show that co-targeting two GPCRs, the vasopressin V2 receptor (V2R) and the calcium sensing receptor, using the novel V2R antagonist lixivaptan in combination with the calcimimetic R-568, reduced cyst progression in two animal models of human PKD. Lixivaptan is expected to have a safer liver profile compared to tolvaptan, the only drug approved to delay PKD progression, based on computational model results and initial clinical evidence. PCK rat and Pkd1RC/RC mouse littermates were fed without or with lixivaptan (0.5%) and R-568 (0.025% for rats and 0.04% for mice), alone or in combination, for 7 (rats) or 13 (mice) weeks. In PCK rats, the combined treatment strongly decreased kidney weight, cyst and fibrosis volumes by 20%, 49%, and 73%, respectively, compared to untreated animals. In Pkd1RC/RC mice, the same parameters were reduced by 20%, 56%, and 69%, respectively. In both cases the combined treatment appeared nominally more effective than the individual drugs used alone. These data point to an intriguing new application for two existing drugs in PKD treatment. The potential for synergy between these two compounds suggested in these animal studies, if confirmed in appropriate clinical investigations, would represent a welcome advancement in the treatment of ADPKD.


Subject(s)
Benzamides/pharmacology , Phenethylamines/pharmacology , Polycystic Kidney, Autosomal Dominant/drug therapy , Propylamines/pharmacology , Pyrroles/pharmacology , Receptors, Calcium-Sensing/antagonists & inhibitors , Receptors, Vasopressin/chemistry , Animals , Cyclic AMP , Drug Therapy, Combination , Female , Male , Mice , Mice, Inbred C57BL , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , Rats , Rats, Sprague-Dawley
9.
Bioorg Chem ; 116: 105301, 2021 11.
Article in English | MEDLINE | ID: mdl-34492558

ABSTRACT

A combination of several pharmacophores in one molecule has been successfully used for multi-target-directed ligands (MTDL) design. New propargylamine substituted derivatives combined with salicylic and cinnamic scaffolds were designed and synthesized as potential cholinesterases and monoamine oxidases (MAOs) inhibitors. They were evaluated invitro for inhibition of acetyl- (AChE) and butyrylcholinesterase (BuChE) using Ellman's method. All the compounds act as dual inhibitors. Most of the derivatives are stronger inhibitors of AChE, the best activity showed 5-bromo-N-(prop-2-yn-1-yl)salicylamide 1e (IC50 = 8.05 µM). Carbamates (4-bromo-2-[(prop-2-yn-1-yl)carbamoyl]phenyl ethyl(methyl)carbamate 2d and 2,4-dibromo-6-[(prop-2-yn-1-yl)carbamoyl]phenyl ethyl(methyl)carbamate 2e were selective and the most active for BuChE (25.10 and 26.09 µM). 4-Bromo-2-[(prop-2-yn-1-ylimino)methyl]phenol 4a was the most potent inhibitor of MAOs (IC50 of 3.95 and ≈10 µM for MAO-B and MAO-A, respectively) along with a balanced inhibition of both cholinesterases being a real MTDL. The mechanism of action was proposed, and binding modes of the hits were studied by molecular docking on human enzymes. Some of the derivatives also exhibited antioxidant properties. Insilico prediction of physicochemical parameters affirm that the molecules would be active after oral administration and able to reach brain tissue.


Subject(s)
Antioxidants/pharmacology , Cholinesterase Inhibitors/pharmacology , Molecular Docking Simulation , Monoamine Oxidase Inhibitors/pharmacology , Pargyline/analogs & derivatives , Propylamines/pharmacology , Animals , Antioxidants/chemical synthesis , Antioxidants/chemistry , Butyrylcholinesterase/metabolism , Cells, Cultured , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Cholinesterases/metabolism , Dose-Response Relationship, Drug , Electrophorus , Hepatocytes/drug effects , Hepatocytes/metabolism , Horses , Humans , Male , Molecular Structure , Monoamine Oxidase/metabolism , Monoamine Oxidase Inhibitors/chemical synthesis , Monoamine Oxidase Inhibitors/chemistry , Pargyline/chemical synthesis , Pargyline/chemistry , Pargyline/pharmacology , Propylamines/chemical synthesis , Propylamines/chemistry , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Structure-Activity Relationship
10.
J Biol Chem ; 297(2): 100940, 2021 08.
Article in English | MEDLINE | ID: mdl-34237302

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 envelope protein (S2-E) is a conserved membrane protein that is important for coronavirus (CoV) assembly and budding. Here, we describe the recombinant expression and purification of S2-E in amphipol-class amphipathic polymer solutions, which solubilize and stabilize membrane proteins, but do not disrupt membranes. We found that amphipol delivery of S2-E to preformed planar bilayers results in spontaneous membrane integration and formation of viroporin cation channels. Amphipol delivery of the S2-E protein to human cells results in plasma membrane integration, followed by retrograde trafficking to the trans-Golgi network and accumulation in swollen perinuclear lysosomal-associated membrane protein 1-positive vesicles, likely lysosomes. CoV envelope proteins have previously been proposed to manipulate the luminal pH of the trans-Golgi network, which serves as an accumulation station for progeny CoV particles prior to cellular egress via lysosomes. Delivery of S2-E to cells will enable chemical biological approaches for future studies of severe acute respiratory syndrome coronavirus 2 pathogenesis and possibly even development of "Trojan horse" antiviral therapies. Finally, this work also establishes a paradigm for amphipol-mediated delivery of membrane proteins to cells.


Subject(s)
Cell Membrane/drug effects , Coronavirus Envelope Proteins/metabolism , Polymers/pharmacology , Propylamines/pharmacology , Surface-Active Agents/pharmacology , trans-Golgi Network/metabolism , Cell Membrane/metabolism , Coronavirus Envelope Proteins/genetics , HeLa Cells , Humans , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Lysosomes/metabolism , Polymers/chemistry , Propylamines/chemistry , Protein Transport , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Surface-Active Agents/chemistry
11.
Am J Nurs ; 121(7): 24, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34156375

ABSTRACT

A rising number of cases of misuse and abuse of propylhexedrine (Benzedrex), an over-the-counter nasal decongestant, have been documented. Misuse of this drug can lead to serious and potentially fatal cardiac and psychiatric adverse effects.


Subject(s)
Nasal Decongestants/adverse effects , Propylamines/adverse effects , Humans , Nasal Decongestants/administration & dosage , Nonprescription Drugs/adverse effects , Propylamines/administration & dosage , Propylamines/pharmacology
12.
Int J Mol Sci ; 22(11)2021 May 21.
Article in English | MEDLINE | ID: mdl-34064275

ABSTRACT

Sigma-1 receptor (chaperone Sigma1R) is an intracellular protein with chaperone functions, which is expressed in various organs, including the brain. Sigma1R participates in the regulation of physiological mechanisms of anxiety (Su, T. P. et al., 2016) and reactions to emotional stress (Hayashi, T., 2015). In 2006, fabomotizole (ethoxy-2-[2-(morpholino)-ethylthio]benzimidazole dihydrochloride) was registered in Russia as an anxiolytic (Seredenin S. and Voronin M., 2009). The molecular targets of fabomotizole are Sigma1R, NRH: quinone reductase 2 (NQO2), and monoamine oxidase A (MAO-A) (Seredenin S. and Voronin M., 2009). The current study aimed to clarify the dependence of fabomotizole anxiolytic action on its interaction with Sigma1R and perform a docking analysis of fabomotizole interaction with Sigma1R. An elevated plus maze (EPM) test revealed that the anxiolytic-like effect of fabomotizole (2.5 mg/kg i.p.) administered to male BALB/c mice 30 min prior EPM exposition was blocked by Sigma1R antagonists BD-1047 (1.0 mg/kg i.p.) and NE-100 (1.0 mg/kg i.p.) pretreatment. Results of initial in silico study showed that fabomotizole locates in the active center of Sigma1R, reproducing the interactions with the site's amino acids common for established Sigma1R ligands, with the ΔGbind value closer to that of agonist (+)-pentazocine in the 6DK1 binding site.


Subject(s)
Anti-Anxiety Agents/pharmacology , Anxiety/drug therapy , Benzimidazoles/pharmacology , Molecular Chaperones/metabolism , Morpholines/pharmacology , Receptors, sigma/metabolism , Animals , Anisoles/pharmacology , Binding Sites/physiology , Brain/drug effects , Brain/metabolism , Ethylenediamines/pharmacology , Ligands , Male , Mice , Mice, Inbred BALB C , Propylamines/pharmacology , Russia , Sigma-1 Receptor
13.
Bioorg Med Chem Lett ; 45: 128135, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34044119

ABSTRACT

Twenty six propargylamine mycophenolate analogues were designed and synthesized from mycophenolic acid 1 employing a key step A3-coupling reaction. Their cytotoxic activity was examined against six cancer cell lines. Compounds 6a, 6j, 6t, 6u, and 6z exhibited selective cytotoxicity towards neuroblastoma (SH-SY5Y) cancer cells and were less toxic to normal cells in comparison to the lead compound, MPA 1 and a standard drug, ellipticine. Molecular docking results suggested that compound 6a is fit well in the key amino acid of three proteins (CDK9, EGFR, and VEGFR-2) as targets in cancer therapy. The propargylamine mycophenolate scaffold might be a valuable starting point for development of new neuroblastoma anticancer drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Mycophenolic Acid/pharmacology , Neuroblastoma/drug therapy , Pargyline/analogs & derivatives , Propylamines/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Docking Simulation , Molecular Structure , Mycophenolic Acid/chemical synthesis , Mycophenolic Acid/chemistry , Neuroblastoma/pathology , Pargyline/chemical synthesis , Pargyline/chemistry , Pargyline/pharmacology , Propylamines/chemical synthesis , Propylamines/chemistry , Structure-Activity Relationship
14.
Brain Res ; 1765: 147508, 2021 08 15.
Article in English | MEDLINE | ID: mdl-33930376

ABSTRACT

Cerebral vasospasm (CVS) causes mortality and morbidity in patients after subarachnoid hemorrhage (SAH). The mechanism and adequate treatment of CVS are still elusive. R-568 is a calcimimetic agent known to exert a vasodilating effect. However, there is no report on its vasodilator effect against SAH-induced vasospasm. In the present study, we investigated the therapeutic effect of R-568 on the SAH-induced CVS model in rats. Seventy-two adult male Sprague-Dawley rats were divided into 8 groups: sham surgery; SAH only; SAH + Vehicle, SAH + R-568; SAH + R-568 + Wortmannin (the PI3K inhibitor); SAH + Wortmannin; SAH + R-568 + Calhex-231 (a calcilytic agent); SAH + Calhex-231. SAH was induced by blood (0.3 mL) given by intracisternal injection. R-568 (20 µM) was administered intracisternal immediately prior to experimental SAH. Basilar arteries (BAs) were obtained to evaluate PI3K/Akt/eNOS pathway (immunoblotting) and morphological changes 48 h after SAH. Perimeters of BAs were decreased by 24.1% in the SAH group compared to the control group and the wall thickness was increased by 75.3%. With R-568 treatment, those percentages were 9.6% and 29.6%, respectively, indicating that vasospasm was considerably improved when compared with the SAH group (P < 0.001 in both). While p-PI3K/PI3K and p-Akt/Akt ratio and eNOS protein expression were markedly decreased in the SAH rats, treatment with R-568 resulted in a significant increase in these levels. The beneficial effects of R-568 were partially blocked in the presence of Calhex-231 and completely blocked in the presence of Wortmannin. Herein, we found that treatment with R-568 would attenuate SAH-induced CVS through the PI3K/Akt/eNOS pathway and demonstrate therapeutic promise in CVS treatment following SAH.


Subject(s)
Phenethylamines/pharmacology , Propylamines/pharmacology , Subarachnoid Hemorrhage/drug therapy , Vasospasm, Intracranial/drug therapy , Animals , Calcimimetic Agents/pharmacology , Disease Models, Animal , Male , Nitric Oxide Synthase Type III/metabolism , Phenethylamines/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Propylamines/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Subarachnoid Hemorrhage/physiopathology , Vasospasm, Intracranial/metabolism
15.
Molecules ; 26(6)2021 Mar 11.
Article in English | MEDLINE | ID: mdl-33799713

ABSTRACT

Osteomyelitis and orthopedic infections are major clinical problems, limited by a lack of antibiotics specialized for such applications. In this paper, we describe the design and synthesis of a novel bone-binding antibiotic (BBA-1) and its subsequent structural and functional characterization. The synthesis of BBA-1 was the result of a two-step chemical conjugation of cationic selective antimicrobial-90 (CSA-90) and the bisphosphonate alendronate (ALN) via a heterobifunctional linker. This was analytically confirmed by HPLC, FT-IR, MS and NMR spectroscopy. BBA-1 showed rapid binding and high affinity to bone mineral in an in vitro hydroxyapatite binding assay. Kirby-Baur assays confirmed that BBA-1 shows a potent antibacterial activity against Staphylococcus aureus and methicillin-resistant S. aureus comparable to CSA-90. Differentiation of cultured osteoblasts in media supplemented with BBA-1 led to increased alkaline phosphatase expression, which is consistent with the pro-osteogenic activity of CSA-90. Bisphosphonates, such as ALN, are inhibitors of protein prenylation, however, the amine conjugation of ALN to CSA-90 disrupted this activity in an in vitro protein prenylation assay. Overall, these findings support the antimicrobial, bone-binding, and pro-osteogenic activities of BBA-1. The compound and related agents have the potential to ensure lasting activity against osteomyelitis after systemic delivery.


Subject(s)
Alendronate/chemistry , Anti-Bacterial Agents/chemical synthesis , Osteomyelitis/drug therapy , Pregnanes/chemistry , Propylamines/chemistry , 3T3 Cells , Alendronate/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Anti-Infective Agents/pharmacology , Bone and Bones/drug effects , Calcification, Physiologic/drug effects , Cell Differentiation/drug effects , Cells, Cultured , Diphosphonates/chemistry , Diphosphonates/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Mice , Osteoblasts/drug effects , Osteogenesis/drug effects , Pregnanes/pharmacology , Propylamines/pharmacology , Spectroscopy, Fourier Transform Infrared , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects
16.
FASEB J ; 35(5): e21581, 2021 05.
Article in English | MEDLINE | ID: mdl-33871072

ABSTRACT

Synaptic plasticity damages play a crucial role in the onset and development of depression, especially in the hippocampus, which is more susceptible to stress and the most frequently studied brain region in depression. And, mitochondria have a major function in executing the complex processes of neurotransmission and plasticity. We have previously demonstrated that Iptakalim (Ipt), a new ATP-sensitive potassium (K-ATP) channel opener, could improve the depressive-like behavior in mice. But the underlying mechanisms are not well understood. The present study demonstrated that Ipt reversed depressive-like phenotype in vivo (chronic mild stress-induced mice model of depression) and in vitro (corticosterone-induced cellular model). Further study showed that Ipt could upregulate the synaptic-related proteins postsynaptic density 95 (PSD 95) and synaptophysin (SYN), and alleviated the synaptic structure damage. Moreover, Ipt could reverse the abnormal mitochondrial fission and fusion, as well as the reduced mitochondrial ATP production and collapse of mitochondrial membrane potential in depressive models. Knocking down the mitochondrial ATP-sensitive potassium (Mito-KATP) channel subunit MitoK partly blocked the above effects of Ipt. Therefore, our results reveal that Ipt can alleviate the abnormal mitochondrial dynamics and function depending on MitoK, contributing to improve synaptic plasticity and exert antidepressive effects. These findings provide a candidate compound and a novel target for antidepressive therapy.


Subject(s)
Depression/drug therapy , KATP Channels/antagonists & inhibitors , Mitochondria/drug effects , Propylamines/pharmacology , Stress, Psychological/complications , Synapses/drug effects , Animals , Depression/etiology , Depression/pathology , Hippocampus/drug effects , Hippocampus/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Neuronal Plasticity , Synapses/metabolism
17.
Int J Mol Sci ; 22(4)2021 Feb 18.
Article in English | MEDLINE | ID: mdl-33670622

ABSTRACT

The Hippo pathway is involved in human tumorigenesis and tissue repair. Here, we investigated the Hippo coactivator Yes-associated protein 1 (YAP1) and the kinase large tumor suppressor 1/2 (LATS1/2) in tumors of the parathyroid glands, which are almost invariably associated with primary hyperparathyroidism. Compared with normal parathyroid glands, parathyroid adenomas (PAds) and carcinomas show variably but reduced nuclear YAP1 expression. The kinase LATS1/2, which phosphorylates YAP1 thus promoting its degradation, was also variably reduced in PAds. Further, YAP1 silencing reduces the expression of the key parathyroid oncosuppressor multiple endocrine neoplasia type 1(MEN1), while MEN1 silencing increases YAP1 expression. Treatment of patient-derived PAds-primary cell cultures and Human embryonic kidney 293A (HEK293A) cells expressing the calcium-sensing receptor (CASR) with the CASR agonist R568 induces YAP1 nuclear accumulation. This effect was prevented by the incubation of the cells with RhoA/Rho-associated coiled-coil-containing protein kinase (ROCK) inhibitors Y27632 and H1152. Lastly, CASR activation increased the expression of the YAP1 gene targets CYR61, CTGF, and WNT5A, and this effect was blunted by YAP1 silencing. Concluding, here we provide preliminary evidence of the involvement of the Hippo pathway in human tumor parathyroid cells and of the existence of a CASR-ROCK-YAP1 axis. We propose a tumor suppressor role for YAP1 and LATS1/2 in parathyroid tumors.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Parathyroid Glands/metabolism , Parathyroid Neoplasms/genetics , Receptors, Calcium-Sensing/genetics , Transcription Factors/genetics , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Adaptor Proteins, Signal Transducing/metabolism , Amides/pharmacology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Gene Expression/drug effects , HEK293 Cells , Humans , Parathyroid Neoplasms/metabolism , Phenethylamines/pharmacology , Propylamines/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Pyridines/pharmacology , RNA Interference , Receptors, Calcium-Sensing/agonists , Receptors, Calcium-Sensing/metabolism , Transcription Factors/metabolism , Tumor Cells, Cultured , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , YAP-Signaling Proteins , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
18.
J Biomed Mater Res A ; 109(5): 713-721, 2021 05.
Article in English | MEDLINE | ID: mdl-32654391

ABSTRACT

The prevalence of periodontal disease poses a significant global health burden. Treatments for these diseases, primarily focused on removal and eradication of dental plaque biofilms, are challenging due to limited access to periodontal pockets where these oral pathogens reside. Herein, we report on the development and characterization of nitric oxide (NO)-releasing carboxymethylcellulose (CMC) derivatives and evaluate their in vitro bactericidal efficacy against planktonic Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans, two prominent periodontopathogens. Bactericidal exposure assays revealed that three of the synthesized NO-releasing polymers were capable of reducing bacterial viability of both species by 99.9% in 2 hr at concentrations of 4 mg ml-1 or lower, reflecting NO's potent and rapid bactericidal action. The NO-releasing CMCs elicited minimal toxicity to human gingival fibroblasts at their bactericidal concentrations following 24-hr exposure.


Subject(s)
Aggregatibacter actinomycetemcomitans/drug effects , Anti-Bacterial Agents/pharmacology , Azo Compounds/pharmacology , Carboxymethylcellulose Sodium , Ethanolamines/pharmacology , Nitric Oxide/pharmacology , Periodontal Diseases/microbiology , Polyamines/pharmacology , Porphyromonas gingivalis/drug effects , Propylamines/pharmacology , Anti-Bacterial Agents/administration & dosage , Azo Compounds/administration & dosage , Azo Compounds/chemistry , Biopolymers , Cell Line , Diamines/chemistry , Drug Carriers , Drug Delivery Systems , Ethanolamines/administration & dosage , Ethanolamines/chemistry , Fibroblasts/drug effects , Gingiva/cytology , Humans , Molecular Structure , Nitric Oxide/administration & dosage , Nitric Oxide/toxicity , Polyamines/administration & dosage , Polyamines/chemistry , Propylamines/administration & dosage , Propylamines/chemistry , Species Specificity , Viscosity
19.
Front Endocrinol (Lausanne) ; 12: 743517, 2021.
Article in English | MEDLINE | ID: mdl-35095753

ABSTRACT

Objective: We identified a novel inactivating mutation in the calcium-sensing receptor (CaSR) gene in a patient with refractory hypocalciuric hypercalcemia and analyzed its function. The effectiveness of radiofrequency ablation of the parathyroid glands to treat hypercalcemia caused by this mutation was explored. Methods: Clinical data of patients before and after radiofrequency ablation were retrospectively analyzed. The CaSR mutation (D99N) found in the patient was studied in cell lines. HEK-293 cells were transfected with plasmids containing wild-type (WT) or mutant CaSR genes (D99N and W718X). Expression levels of the respective CaSR proteins were measured, and their functions were assessed by examining the effect of NPS R-568 (a CaSR agonist) on intracellular Ca2+ oscillations and that of exogenous parathyroid hormone (PTH) on intracellular cyclic adenosine monophosphate (cAMP) levels. Results: The effectiveness of pharmacological treatment was poor, whereas radiofrequency ablation of the parathyroid glands resulted in controlled blood calcium and PTH levels in the patient. In cell lines, upon NPS R-568 administration, the amplitude of intracellular Ca2+ oscillations in the D99N group was lower than that in the WT group and higher than that in the W718X group. Upon administration of PTH, intracellular cAMP levels in the D99N group were higher than those in the WT group and lower than those in the W718X group. Conclusion: The homozygous mutation D99N reduced CaSR activity and caused more severe hypocalciuric hypercalcemia. For patients with this type of hypercalcemia and poor response to pharmacological treatments, radiofrequency ablation of the parathyroid glands may be a suitable treatment option.


Subject(s)
Hypercalcemia/surgery , Parathyroid Glands/surgery , Radiofrequency Ablation/methods , Receptors, Calcium-Sensing/genetics , Adult , Calcium/metabolism , Calcium-Regulating Hormones and Agents/therapeutic use , Cinacalcet/therapeutic use , HEK293 Cells , Humans , Hypercalcemia/blood , Male , Mutation , Parathyroid Glands/metabolism , Parathyroid Hormone/blood , Phenethylamines/pharmacology , Propylamines/pharmacology , Receptors, Calcium-Sensing/metabolism , Treatment Failure
20.
J Biomater Appl ; 35(8): 1019-1033, 2021 03.
Article in English | MEDLINE | ID: mdl-33290123

ABSTRACT

Tacrolimus (TAC), a potent immunosuppressive macrolide, has been investigated for ocular diseases due to promising results in the treatment of anterior and posterior segments eye diseases. Mesoporous and functionalized silica nanoparticles show potential as TAC delivery platforms owing to their interesting characteristic as large surface area, uniform pore size distribution, high pore volume, and excellent biocompatibility. The purpose of this study was to incorporate TAC in functionalized silica nanoparticles with 3-aminopropyltriethoxysilane (MSNAPTES) and investigate the safety and biocompatibility of the systems. The MSNAPTES and MSNAPTES TAC nanoparticles were characterized. The in vitro cytotoxicity of MSNAPTES and MSNAPTES load with TAC (MSNAPTES-TAC) in retinal pigment epithelial cells (ARPE-19) was determined, chorioallantoic membrane (CAM) assay model was used to investigate the in vivo biocompatibility, and safety of intravitreal injection was evaluated using clinical examination (assessment of intraocular pressure and indirect fundus ophthalmoscopy), electroretinographic (ERG) and histologic studies in rats' eyes. The elemental analysis (CHN), thermogravimetric (TGA), photon correlation spectroscopy and Fourier transform infrared (FTIR) analysis confirmed the presence of functionalized agent and TAC in the MSNAPTES nanoparticles. TAC loading was estimated at 7% for the MSNAPTES TAC nanoparticles. MSNAPTES and MSNAPTES TAC did not present in vitro cytotoxicity. The drug delivery systems showed good biocompatibility on CAM. No retinal abnormalities, vitreous hemorrhage, neovascularization, retinal detachment, and optic nerve atrophy were observed during the in vivo study. Follow-up ERGs showed no changes in the function of the retina cells after 15 days of intravitreal injection, and histopathologic observations support these findings. In conclusion, MSNAPTES TAC was successfully synthesized, and physicochemical analyses confirmed the presence of TAC in the nanoparticles. In vitro and in vivo studies indicated that MSNAPTES TAC was safe to intravitreal administration. Taking into account the enormous potential of MSNAPTES to carry TAC, this platform could be a promising strategy for TAC ocular drug delivery in the treatment of eye diseases.


Subject(s)
Nanoparticles/chemistry , Silicon Dioxide/chemistry , Tacrolimus/administration & dosage , Administration, Intravesical , Animals , Biocompatible Materials/administration & dosage , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Line , Cell Survival/drug effects , Drug Delivery Systems , Humans , Nanoparticles/administration & dosage , Particle Size , Porosity , Propylamines/administration & dosage , Propylamines/chemistry , Propylamines/pharmacology , Rats , Silanes/administration & dosage , Silanes/chemistry , Silanes/pharmacology , Silicon Dioxide/administration & dosage , Silicon Dioxide/pharmacology , Tacrolimus/chemistry , Tacrolimus/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL