Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38861338

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic lung disease that leads to respiratory decline due to scarring and thickening of lung tissues. Multiple pathways contribute to the fibrotic process in this disease, such as inflammation, epithelial to mesenchymal transition and oxidative stress. The RhoA/ROCK signaling pathway is a key regulator of profibrotic signaling, as it affects the organization of actin-myosin and the remodeling of the extracellular matrix. ROCK1/2, a downstream effector of RhoA, is overexpressed in IPF patients and is a promising target for IPF therapy. However, due to hypotensive side effects of ROCK1/2 inhibitors, selective ROCK2 compounds are being explored. In this study, we report the discovery of GNS-3595, a potent and selective ROCK2 inhibitor that has ~80-fold selectivity over ROCK1 at physiological concentrations of ATP. GNS-3595 effectively inhibited ROCK2-mediated phosphorylation of myosin light chain (p-MLC) and reduced the expression of fibrosis-related proteins, such as collagen, fibronectin, and alpha-smooth muscle actin (α-SMA) in various in vitro cellular models. GNS-3595 also prevented transforming growth factor beta (TGF-ß)-induced fibroblast-to-myofibroblast transition (FMT). Additionally, in a bleomycin-induced mouse model of pulmonary fibrosis, therapeutic exposure to GNS-3595, suppressed lung fibrosis, stabilized body weight loss, and prevented fibrosis-induced lung weight gain. Transcriptome and protein expression analysis from lung tissues showed that GNS-3595 can revert the fibrosis-related gene expression induced by bleomycin. These results indicate that GNS-3595 is a highly potent, selective, and orally active ROCK2 inhibitor with promising therapeutic efficacy against pulmonary fibrosis.

2.
Protein Sci ; 29(9): 1902-1910, 2020 09.
Article in English | MEDLINE | ID: mdl-32643196

ABSTRACT

Human immunodeficiency virus (HIV)-1 remains as a global health issue that is primarily treated with highly active antiretroviral therapy, a combination of drugs that target the viral life cycle. One class of these drugs are non-nucleoside reverse transcriptase inhibitors (NNRTIs) that target the viral reverse transcriptase (RT). First generation NNRTIs were troubled with poor pharmacological properties and drug resistance, incentivizing the development of improved compounds. One class of developed compounds are the 2-naphthyl phenyl ethers, showing promising efficacy against the Y181C RT mutation. Further biochemical and structural work demonstrated differences in potency against the Y181C mutation and binding mode of the compounds. This work aims to understand the relationship between the binding mode and ability to overcome drug resistance using macromolecular x-ray crystallography. Comparison of 2-naphthyl phenyl ethers bound to Y181C RT reveal that compounds that interact with the invariant W229 are more capable of retaining efficacy against the resistance mutation. Additional modifications to these compounds at the 4-position, computationally designed to compensate for the Y181C mutation, do not demonstrate improved potency. Ultimately, we highlight important considerations for the development of future HIV-1 drugs that are able to combat drug resistance.


Subject(s)
HIV Reverse Transcriptase/antagonists & inhibitors , HIV Reverse Transcriptase/chemistry , HIV-1/enzymology , Mutation, Missense , Phenyl Ethers/chemistry , Reverse Transcriptase Inhibitors/chemistry , Amino Acid Substitution , Binding Sites , HIV Reverse Transcriptase/genetics , HIV-1/genetics
3.
Bioorg Med Chem Lett ; 29(16): 2182-2188, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31281023

ABSTRACT

The development of efficacious NNRTIs for HIV/AIDS therapy is commonly met with the emergence of drug resistant strains, including the Y181C variant. Using a computationally-guided approach, we synthesized the catechol diether series of NNRTIs, which display sub-nanomolar potency in cellular assays. Among the most potent were a series of 2-cyanoindolizine substituted catechol diethers, including Compound 1. We present here a thorough evaluation of this compound, including biochemical, cellular, and structural studies. The compound demonstrates low nanomolar potency against both WT and Y181C HIV-1 RT in in vitro and cellular assays. Our crystal structures of both the wildtype and mutant forms of RT in complex with Compound 1 allow the interrogation of this compound's features that allow it to maintain strong efficacy against the drug resistant mutant. Among these are compensatory shifts in the NNRTI binding pocket, persistence of multiple hydrogen bonds, and van der Waals contacts throughout the binding site. Further, the fluorine at the C6 position of the indolizine moiety makes multiple favorable interactions with both RT forms. The present study highlights the indolizine-substituted catechol diether class of NNRTIs as promising therapeutic candidates possessing optimal pharmacological properties and significant potency against multiple RT variants.


Subject(s)
Anti-HIV Agents/therapeutic use , Catechols/chemistry , HIV Reverse Transcriptase/metabolism , Reverse Transcriptase Inhibitors/therapeutic use , Anti-HIV Agents/pharmacology , Drug Design , Molecular Structure , Reverse Transcriptase Inhibitors/pharmacology
5.
Proc Natl Acad Sci U S A ; 115(4): E802-E811, 2018 01 23.
Article in English | MEDLINE | ID: mdl-29279368

ABSTRACT

The HIV-1 pandemic affecting over 37 million people worldwide continues, with nearly one-half of the infected population on highly active antiretroviral therapy (HAART). Major therapeutic challenges remain because of the emergence of drug-resistant HIV-1 strains, limitations because of safety and toxicity with current HIV-1 drugs, and patient compliance for lifelong, daily treatment regimens. Nonnucleoside reverse transcriptase inhibitors (NNRTIs) that target the viral polymerase have been a key component of the current HIV-1 combination drug regimens; however, these issues hamper them. Thus, the development of novel more effective NNRTIs as anti-HIV-1 agents with fewer long-term liabilities, efficacy on new drug-resistant HIV-1 strains, and less frequent dosing is crucial. Using a computational and structure-based design strategy to guide lead optimization, a 5 µM virtual screening hit was transformed to a series of very potent nanomolar to picomolar catechol diethers. One representative, compound I, was shown to have nanomolar activity in HIV-1-infected T cells, potency on clinically relevant HIV-1 drug-resistant strains, lack of cytotoxicity and off-target effects, and excellent in vivo pharmacokinetic behavior. In this report, we show the feasibility of compound I as a late-stage preclinical candidate by establishing synergistic antiviral activity with existing HIV-1 drugs and clinical candidates and efficacy in HIV-1-infected humanized [human peripheral blood lymphocyte (Hu-PBL)] mice by completely suppressing viral loads and preventing human CD4+ T-cell loss. Moreover, a long-acting nanoformulation of compound I [compound I nanoparticle (compound I-NP)] in poly(lactide-coglycolide) (PLGA) was developed that shows sustained maintenance of plasma drug concentrations and drug efficacy for almost 3 weeks after a single dose.


Subject(s)
Anti-HIV Agents/administration & dosage , Drug Delivery Systems , HIV Infections/drug therapy , HIV-1 , Animals , Anti-HIV Agents/pharmacokinetics , Computer Simulation , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Synergism , Mice , Mice, Inbred BALB C , Nanoparticles
6.
Proc Natl Acad Sci U S A ; 114(36): 9725-9730, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28827354

ABSTRACT

Development of resistance remains a major challenge for drugs to treat HIV-1 infections, including those targeting the essential viral polymerase, HIV-1 reverse transcriptase (RT). Resistance associated with the Tyr181Cys mutation in HIV-1 RT has been a key roadblock in the discovery of nonnucleoside RT inhibitors (NNRTIs). It is the principal point mutation that arises from treatment of HIV-infected patients with nevirapine, the first-in-class drug still widely used, especially in developing countries. We report covalent inhibitors of Tyr181Cys RT (CRTIs) that can completely knock out activity of the resistant mutant and of the particularly challenging Lys103Asn/Tyr181Cys variant. Conclusive evidence for the covalent modification of Cys181 is provided from enzyme inhibition kinetics, mass spectrometry, protein crystallography, and antiviral activity in infected human T-cell assays. The CRTIs are also shown to be selective for Cys181 and have lower cytotoxicity than the approved NNRTI drugs efavirenz and rilpivirine.


Subject(s)
Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , HIV Reverse Transcriptase/antagonists & inhibitors , HIV Reverse Transcriptase/chemistry , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Amino Acid Substitution , Anti-HIV Agents/chemical synthesis , Cell Line , Crystallography, X-Ray , Drug Design , Drug Resistance, Viral/genetics , HIV Reverse Transcriptase/genetics , HIV-1/drug effects , HIV-1/enzymology , HIV-1/genetics , Humans , Kinetics , Models, Molecular , Point Mutation , Protein Conformation , Reverse Transcriptase Inhibitors/chemical synthesis , Spectrometry, Mass, Electrospray Ionization
7.
Mol Pharmacol ; 91(4): 383-391, 2017 04.
Article in English | MEDLINE | ID: mdl-28167742

ABSTRACT

The clinical benefits of HIV-1 non-nucleoside reverse transcriptase (RT) inhibitors (NNRTIs) are hindered by their unsatisfactory pharmacokinetic (PK) properties along with the rapid development of drug-resistant variants. However, the clinical efficacy of these inhibitors can be improved by developing compounds with enhanced pharmacological profiles and heightened antiviral activity. We used computational and structure-guided design to develop two next-generation NNRTI drug candidates, compounds I and II, which are members of a class of catechol diethers. We evaluated the preclinical potential of these compounds in BALB/c mice because of their high solubility (510 µg/ml for compound I and 82.9 µg/ml for compound II), low cytotoxicity, and enhanced antiviral activity against wild-type (WT) HIV-1 RT and resistant variants. Additionally, crystal structures of compounds I and II with WT RT suggested an optimal binding to the NNRTI binding pocket favoring the high anti-viral potency. A single intraperitoneal dose of compounds I and II exhibited a prolonged serum residence time of 48 hours and concentration maximum (Cmax) of 4000- to 15,000-fold higher than their therapeutic/effective concentrations. These Cmax values were 4- to 15-fold lower than their cytotoxic concentrations observed in MT-2 cells. Compound II showed an enhanced area under the curve (0-last) and decreased plasma clearance over compound I and efavirenz, the standard of care NNRTI. Hence, the overall (PK) profile of compound II was excellent compared with that of compound I and efavirenz. Furthermore, both compounds were very well tolerated in BALB/c mice without any detectable acute toxicity. Taken together, these data suggest that compounds I and II possess improved anti-HIV-1 potency, remarkable in vivo safety, and prolonged in vivo circulation time, suggesting strong potential for further development as new NNRTIs for the potential treatment of HIV infection.


Subject(s)
Drug Design , Drug Evaluation, Preclinical , HIV Infections/drug therapy , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/therapeutic use , Alkynes , Animals , Benzoxazines/chemistry , Benzoxazines/pharmacology , Crystallography, X-Ray , Cyclopropanes , Female , HIV Infections/virology , HIV-1/drug effects , Humans , Inhibitory Concentration 50 , Mice, Inbred BALB C , Reverse Transcriptase Inhibitors/pharmacokinetics , Reverse Transcriptase Inhibitors/toxicity , Solubility
8.
ACS Med Chem Lett ; 7(12): 1156-1160, 2016 Dec 08.
Article in English | MEDLINE | ID: mdl-27994756

ABSTRACT

Catechol diethers that incorporate a 7-cyano-2-naphthyl substituent are reported as non-nucleoside inhibitors of HIV-1 reverse transcriptase (NNRTIs). Many of the compounds have 1-10 nM potencies toward wild-type HIV-1. An interesting conformational effect allows two unique conformers for the naphthyl group in complexes with HIV-RT. X-ray crystal structures for 4a and 4f illustrate the alternatives.

9.
ACS Med Chem Lett ; 6(10): 1075-9, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26487915

ABSTRACT

Catechol diether compounds have nanomolar antiviral and enzymatic activity against HIV with reverse transcriptase (RT) variants containing K101P, a mutation that confers high-level resistance to FDA-approved non-nucleoside inhibitors efavirenz and rilpivirine. Kinetic data suggests that RT (K101P) variants are as catalytically fit as wild-type and thus can potentially increase in the viral population as more antiviral regimens include efavirenz or rilpivirine. Comparison of wild-type structures and a new crystal structure of RT (K101P) in complex with a leading compound confirms that the K101P mutation is not a liability for the catechol diethers while suggesting that key interactions are lost with efavirenz and rilpivirine.

10.
Bioorg Chem ; 61: 58-65, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26123174

ABSTRACT

The pyridine core skeleton of the previously reported dichloropyridine-based potent hP2X7 receptor antagonist 5 (IC50 = 13 nM in hP2X7-expressing HEK293 cells) was modified with various heterocyclic scaffolds. Among the derivatives with quinoline, quinazoline, acridine, and purine scaffolds, the chloropurine-based analog 9o exhibited the most potent antagonistic activity, with an IC50 value of 176 ± 37 nM in an ethidium bromide uptake assay. In addition, 9o significantly inhibited IL-1ß release in THP-1 cells stimulated with LPS/IFN-γ/BzATP (IC50 = 120 ± 15 nM). Although 9o was less active than the previous antagonist 5, 9o exhibited greatly improved metabolic stability in the in vitro evaluation (71.4% in human, 72.3% in mouse).


Subject(s)
Heterocyclic Compounds/chemistry , Purinergic P2X Receptor Antagonists/chemistry , Purines/chemistry , Animals , Drug Evaluation, Preclinical , HEK293 Cells , Heterocyclic Compounds/chemical synthesis , Humans , Mice , Microsomes, Liver/metabolism , Protein Binding , Purinergic P2X Receptor Antagonists/metabolism , Receptors, Purinergic P2X7/chemistry , Receptors, Purinergic P2X7/metabolism , Structure-Activity Relationship
11.
Bioorg Med Chem Lett ; 25(21): 4824-4827, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26166629

ABSTRACT

Non-nucleoside inhibitors of HIV-1 reverse transcriptase (HIV-RT) are reported that incorporate a 7-indolizinylamino or 2-naphthylamino substituent on a pyrimidine or 1,3,5-triazine core. The most potent compounds show below 10 nanomolar activity towards wild-type HIV-1 and variants bearing Tyr181Cys and Lys103Asn/Tyr181Cys resistance mutations. The compounds also feature good aqueous solubility. Crystal structures for two complexes enhance the analysis of the structure-activity data.


Subject(s)
Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , Azabicyclo Compounds/pharmacology , Bridged Bicyclo Compounds/pharmacology , Drug Discovery , HIV Reverse Transcriptase/antagonists & inhibitors , HIV/drug effects , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/pharmacology , Triazines/pharmacology , Anti-HIV Agents/chemical synthesis , Azabicyclo Compounds/chemical synthesis , Azabicyclo Compounds/chemistry , Bridged Bicyclo Compounds/chemical synthesis , Bridged Bicyclo Compounds/chemistry , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , HIV Reverse Transcriptase/metabolism , Humans , Models, Molecular , Molecular Structure , Reverse Transcriptase Inhibitors/chemical synthesis , Solubility , Structure-Activity Relationship , Triazines/chemical synthesis , Triazines/chemistry
12.
ACS Appl Mater Interfaces ; 7(24): 13405-14, 2015 Jun 24.
Article in English | MEDLINE | ID: mdl-26010272

ABSTRACT

Flexible supercapacitor electrodes have been fabricated by simple fabrication technique using graphite nanoflakes on polymer lapping films as flexible substrate. An additional thin layer of conducting polymer polypyrrole over the electrode improved the surface conductivity and exhibited excellent electrochemical performances. Such capacitor films showed better energy density and power density with a maximum capacitance value of 37 mF cm(-2) in a half cell configuration using 1 M H2SO4 electrolyte, 23 mF cm(-2) in full cell, and 6 mF cm(-2) as planar cell configuration using poly(vinyl alcohol) (PVA)/phosphoric acid (H3PO4) solid state electrolyte. Moreover, the graphite nanoflakes/polypyrrole over polymer lapping film demonstrated good flexibility and cyclic stability.

13.
ACS Med Chem Lett ; 5(11): 1259-62, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25408842

ABSTRACT

Catechol diethers that incorporate a 6-cyano-1-naphthyl substituent have been explored as non-nucleoside inhibitors of HIV-1 reverse transcriptase (NNRTIs). Promising compounds are reported that show midpicomolar activity against the wild-type virus and sub-20 nM activity against viral variants bearing Tyr181Cys and Lys103Asn mutations in HIV-RT. An X-ray crystal structure at 2.49 Å resolution is also reported for the key compound 6e with HIV-RT.

14.
J Am Chem Soc ; 135(44): 16705-13, 2013 Nov 06.
Article in English | MEDLINE | ID: mdl-24151856

ABSTRACT

Members of the catechol diether class are highly potent non-nucleoside inhibitors of HIV-1 reverse transcriptase (NNRTIs). The most active compounds yield EC50 values below 0.5 nM in assays using human T-cells infected by wild-type HIV-1. However, these compounds such as rilpivirine, the most recently FDA-approved NNRTI, bear a cyanovinylphenyl (CVP) group. This is an uncommon substructure in drugs that gives reactivity concerns. In the present work, computer simulations were used to design bicyclic replacements for the CVP group. The predicted viability of a 2-cyanoindolizinyl alternative was confirmed experimentally and provided compounds with 0.4 nM activity against the wild-type virus. The compounds also performed well with EC50 values of 10 nM against the challenging HIV-1 variant that contains the Lys103Asn/Tyr181Cys double mutation in the RT enzyme. Indolyl and benzofuranyl analogues were also investigated; the most potent compounds in these cases have EC50 values toward wild-type HIV-1 near 10 nM and high-nanomolar activities toward the double-variant. The structural expectations from the modeling were much enhanced by obtaining an X-ray crystal structure at 2.88 Å resolution for the complex of the parent 2-cyanoindolizine 10b and HIV-1 RT. The aqueous solubilities of the most potent indolizine analogues were also measured to be ~40 µg/mL, which is similar to that for the approved drug efavirenz and ~1000-fold greater than for rilpivirine.


Subject(s)
Anti-HIV Agents/pharmacology , Bridged Bicyclo Compounds/pharmacology , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , Reverse Transcriptase Inhibitors/pharmacology , Anti-HIV Agents/chemical synthesis , Anti-HIV Agents/chemistry , Bridged Bicyclo Compounds/chemical synthesis , Bridged Bicyclo Compounds/chemistry , Crystallography, X-Ray , Dose-Response Relationship, Drug , HIV Reverse Transcriptase/metabolism , Microbial Sensitivity Tests , Models, Molecular , Molecular Dynamics Simulation , Molecular Structure , Monte Carlo Method , Reverse Transcriptase Inhibitors/chemical synthesis , Reverse Transcriptase Inhibitors/chemistry , Structure-Activity Relationship
15.
Bioorg Med Chem Lett ; 22(22): 6952-6, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23062551

ABSTRACT

Peptidomimetic anti-viral agents against Coxsackievirus B3 (CVB3) were developed using a strategy involving the inhibition of 3C protease (CVB3 3C(pro)), a target for CVB3-mediated myocarditis or pericarditis. In an attempt to improve the inhibitory activity against CVB3, a variety of hetero-aromatic groups were incorporated into the α,ß-unsaturated ester as Michael acceptor moiety, which is the position of interaction with the cysteine moiety in the P1' active site of CVB3 3C(pro). Among these hetero-aromatic groups, the quinoline analogs 9c and 9e, with IC(50) values of 250 and 130 nM as determined from an enzyme assay, significantly inhibited the CVB3-mediated cell cytotoxicity, indicating parallel anti-viral activities. A comparison of the binding modes of the potent inhibitor 9e and the relatively weak inhibitor 9n was explored in a molecular docking study, which revealed that compound 9n lacked hydrogen bonds in its interactions with Gly129, 128, and 145.


Subject(s)
Antiviral Agents/chemistry , Enterovirus/enzymology , Oligopeptides/chemistry , Protease Inhibitors/chemistry , Quinolines/chemistry , Viral Proteins/antagonists & inhibitors , 3C Viral Proteases , Antiviral Agents/chemical synthesis , Antiviral Agents/toxicity , Binding Sites , Catalytic Domain , Cell Survival/drug effects , Cysteine Endopeptidases/metabolism , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Hydrogen Bonding , Molecular Docking Simulation , Oligopeptides/chemical synthesis , Oligopeptides/toxicity , Protease Inhibitors/chemical synthesis , Protease Inhibitors/toxicity , Quinolines/chemical synthesis , Quinolines/toxicity , Viral Proteins/metabolism
16.
J Med Chem ; 55(8): 3687-98, 2012 Apr 26.
Article in English | MEDLINE | ID: mdl-22400713

ABSTRACT

Screening of a library of chemical compounds showed that the dichloropyridine-based analogue 9 was a novel P2X(7) receptor antagonist. To optimize its activity, we assessed the structure-activity relationships (SAR) of 9, focusing on the hydrazide linker, the dichloropyridine skeleton, and the hydrophobic acyl (R(2)) group. We found that the hydrazide linker and the 3,5-disubstituted chlorides in the pyridine skeleton were critical for P2X(7) antagonistic activity and that the presence of hydrophobic polycycloalkyl groups at the R(2) position optimized antagonistic activity. In the EtBr uptake assay in hP2X(7)-expressing HEK293 cells, the optimized antagonists, 51 and 52, had IC(50) values of 4.9 and 13 nM, respectively. The antagonistic effects of 51 and 52 were paralleled by their ability to inhibit the release of the pro-inflammatory cytokine, IL-1ß, by LPS/IFN-γ/BzATP stimulation of THP-1 cells (IC(50) = 1.3 and 9.2 nM, respectively). In addition, 52 strongly inhibited iNOS/COX-2 expression and NO production in THP-1 cells, further indicating that this compound blocks inflammatory signaling and suggesting that the dichloropyridine analogues may be useful in developing P2X(7) receptor targeted anti-inflammatory agents.


Subject(s)
Purinergic P2X Receptor Antagonists/chemistry , Pyridines/pharmacology , Receptors, Purinergic P2X7/drug effects , Cell Line, Tumor , Cyclooxygenase 2/biosynthesis , HEK293 Cells , Humans , Interleukin-18/antagonists & inhibitors , Interleukin-18/metabolism , Nitric Oxide Synthase Type II/biosynthesis , Pyridazines/chemistry , Pyridazines/pharmacology , Pyridines/chemistry , Structure-Activity Relationship
18.
Bioorg Med Chem Lett ; 21(19): 5726-9, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21873054

ABSTRACT

Compounds having α,ß-unsaturated lactones display a variety of biological activities. Many research groups have tested both natural and unnatural α,ß-unsaturated lactones for as-yet undiscovered biological properties. We synthesized α,ß-unsaturated lactones with various substituents at the δ-position and studied their immunosuppressive effects, that is, the inhibition of Interleukin-2 (IL-2) production. Among the compounds synthesized, the benzofuran-substituted α,ß-unsaturated lactone 4h showed the best inhibitory activity toward IL-2 production in Jurkat e6-1 T lymphocytes (IC(50)=66.9 nM) without cytotoxicity at 10 µM. The results indicated that 4h may be useful as a potent immunosuppressive agent, as well as in IL-2-related studies.


Subject(s)
Immunosuppressive Agents/chemical synthesis , Lactones/chemical synthesis , Cytotoxicity Tests, Immunologic , Drug Evaluation, Preclinical , Humans , Immunosuppressive Agents/metabolism , Immunosuppressive Agents/pharmacology , Inflammation/drug therapy , Inhibitory Concentration 50 , Interleukin-2/biosynthesis , Jurkat Cells , Lactones/metabolism , Lactones/pharmacology , Molecular Structure , Structure-Activity Relationship , T-Lymphocytes/metabolism
19.
Toxicol Appl Pharmacol ; 252(2): 192-200, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21320518

ABSTRACT

The P2X(7) receptor (P2X(7)R), a member of the ATP-gated ion channel family, is regarded as a promising target for therapy of immune-related diseases including rheumatoid arthritis and chronic pain. A group of novel protoberberine analogs (compounds 3-5), discovered by screening of chemical libraries, was here investigated with respect to their function as P2X(7)R antagonists. Compounds 3-5 non-competitively inhibited BzATP-induced ethidium ion influx into hP2X(7)-expressing HEK293 cells, with IC(50) values of 100-300nM. This antagonistic action on the channel further confirmed that both BzATP-induced inward currents and Ca(2+) influx were strongly inhibited by compounds 3-5 in patch-clamp and Ca(2+) influx assays. The antagonists also effectively suppressed downstream signaling of P2X(7) receptors including IL-1ß release and phosphorylation of ERK1/2 and p38 proteins in hP2X(7)-expressing HEK293 cells or in differentiated human monocytes (THP-1 cells). Moreover, IL-2 secretion from CD3/CD28-stimulated Jurkat T cell was also dramatically inhibited by the antagonist. These results imply that novel protoberberine analogs may modulate P2X(7) receptor-mediated immune responses by allosteric inhibition of the receptor.


Subject(s)
Berberine Alkaloids/chemistry , Berberine Alkaloids/pharmacology , Purinergic P2 Receptor Antagonists/chemistry , Purinergic P2 Receptor Antagonists/pharmacology , Receptors, Purinergic P2X7 , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Jurkat Cells , Receptors, Purinergic P2X7/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...