Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Channels (Austin) ; 15(1): 179-193, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33427574

RESUMO

A fundamental mechanism that drives the propagation of electrical signals in the nervous system is the activation of voltage-gated sodium channels. The sodium channel subtype Nav1.7 is critical for the transmission of pain-related signaling, with gain-of-function mutations in Nav1.7 resulting in various painful pathologies. Loss-of-function mutations cause complete insensitivity to pain and anosmia in humans that otherwise have normal nervous system function, rendering Nav1.7 an attractive target for the treatment of pain. Despite this, no Nav1.7 selective therapeutic has been approved for use as an analgesic to date. Here we present a summary of research that has focused on engineering peptides found in spider venoms to produce Nav1.7 selective antagonists. We discuss the progress that has been made on various scaffolds from different venom families and highlight the challenges that remain in the effort to produce a Nav1.7 selective, venom-based analgesic.


Assuntos
Venenos de Aranha , Analgésicos , Canal de Sódio Disparado por Voltagem NAV1.7 , Dor
2.
J Clin Invest ; 130(11): 6158-6170, 2020 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-33074244

RESUMO

The α6ß4 nicotinic acetylcholine receptor (nAChR) is enriched in dorsal root ganglia neurons and is an attractive non-opioid therapeutic target for pain. However, difficulty expressing human α6ß4 receptors in recombinant systems has precluded drug discovery. Here, genome-wide screening identified accessory proteins that enable reconstitution of human α6ß4 nAChRs. BARP, an auxiliary subunit of voltage-dependent calcium channels, promoted α6ß4 surface expression while IRE1α, an unfolded protein response sensor, enhanced α6ß4 receptor assembly. Effects on α6ß4 involve BARP's N-terminal region and IRE1α's splicing of XBP1 mRNA. Furthermore, clinical efficacy of nicotinic agents in relieving neuropathic pain best correlated with their activity on α6ß4. Finally, BARP-knockout, but not NACHO-knockout mice lacked nicotine-induced antiallodynia, highlighting the functional importance of α6ß4 in pain. These results identify roles for IRE1α and BARP in neurotransmitter receptor assembly and unlock drug discovery for the previously elusive α6ß4 receptor.


Assuntos
Agonistas Colinérgicos/farmacologia , Endorribonucleases/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Receptores Colinérgicos/biossíntese , Animais , Endorribonucleases/genética , Células HEK293 , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Proteínas Serina-Treonina Quinases/genética , Splicing de RNA/efeitos dos fármacos , Ratos , Receptores Colinérgicos/genética , Proteína 1 de Ligação a X-Box/genética
3.
J Biol Chem ; 295(13): 4359-4366, 2020 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-32079674

RESUMO

Excitatory amino acid transporters (EAATs) represent a protein family that is an emerging drug target with great therapeutic potential for managing central nervous system disorders characterized by dysregulation of glutamatergic neurotransmission. As such, it is of significant interest to discover selective modulators of EAAT2 function. Here, we applied computational methods to identify specific EAAT2 inhibitors. Utilizing a homology model of human EAAT2, we identified a binding pocket at the interface of the transport and trimerization domain. We next conducted a high-throughput virtual screen against this site and identified a selective class of EAAT2 inhibitors that were tested in glutamate uptake and whole-cell electrophysiology assays. These compounds represent potentially useful pharmacological tools suitable for further exploration of the therapeutic potential of EAAT2 and may provide molecular insights into mechanisms of allosteric modulation for glutamate transporters.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/antagonistas & inibidores , Sítios de Ligação/efeitos dos fármacos , Doenças do Sistema Nervoso Central/tratamento farmacológico , Transportador 2 de Aminoácido Excitatório/antagonistas & inibidores , Sistema X-AG de Transporte de Aminoácidos/química , Sistema X-AG de Transporte de Aminoácidos/genética , Animais , Sítios de Ligação/genética , Transporte Biológico/efeitos dos fármacos , Doenças do Sistema Nervoso Central/genética , Doenças do Sistema Nervoso Central/patologia , Biologia Computacional , Transportador 2 de Aminoácido Excitatório/química , Transportador 2 de Aminoácido Excitatório/genética , Humanos , Ligação Proteica/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Interface Usuário-Computador
4.
J Biol Chem ; 295(5): 1315-1327, 2020 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-31871053

RESUMO

Pain is a significant public health burden in the United States, and current treatment approaches rely heavily on opioids, which often have limited efficacy and can lead to addiction. In humans, functional loss of the voltage-gated sodium channel Nav1.7 leads to pain insensitivity without deficits in the central nervous system. Accordingly, discovery of a selective Nav1.7 antagonist should provide an analgesic without abuse liability and an improved side-effect profile. Huwentoxin-IV, a component of tarantula venom, potently blocks sodium channels and is an attractive scaffold for engineering a Nav1.7-selective molecule. To define the functional impact of alterations in huwentoxin-IV sequence, we produced a library of 373 point mutants and tested them for Nav1.7 and Nav1.2 activity. We then combined favorable individual changes to produce combinatorial mutants that showed further improvements in Nav1.7 potency (E1N, E4D, Y33W, Q34S-Nav1.7 pIC50 = 8.1 ± 0.08) and increased selectivity over other Nav isoforms (E1N, R26K, Q34S, G36I, Nav1.7 pIC50 = 7.2 ± 0.1, Nav1.2 pIC50 = 6.1 ± 0.18, Nav1.3 pIC50 = 6.4 ± 1.0), Nav1.4 is inactive at 3 µm, and Nav1.5 is inactive at 10 µm We also substituted noncoded amino acids at select positions in huwentoxin-IV. Based on these results, we identify key determinants of huwentoxin's Nav1.7 inhibition and propose a model for huwentoxin-IV's interaction with Nav1.7. These findings uncover fundamental features of huwentoxin involved in Nav1.7 blockade, provide a foundation for additional optimization of this molecule, and offer a basis for the development of a safe and effective analgesic.


Assuntos
Analgésicos/farmacologia , Canal de Sódio Disparado por Voltagem NAV1.7/efeitos dos fármacos , Venenos de Aranha/química , Venenos de Aranha/genética , Bloqueadores do Canal de Sódio Disparado por Voltagem/farmacologia , Sequência de Aminoácidos/genética , Desenvolvimento de Medicamentos , Células HEK293 , Humanos , Simulação de Acoplamento Molecular , Mutagênese , Canal de Sódio Disparado por Voltagem NAV1.2/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.2/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo , Dor/tratamento farmacológico , Biblioteca de Peptídeos , Mutação Puntual , Engenharia de Proteínas , Isoformas de Proteínas , Proteínas Recombinantes , Venenos de Aranha/isolamento & purificação
5.
J Med Chem ; 60(11): 4559-4572, 2017 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-28493698

RESUMO

The synthesis and preclinical characterization of novel 4-(R)-methyl-6,7-dihydro-4H-triazolo[4,5-c]pyridines that are potent and selective brain penetrant P2X7 antagonists are described. Optimization efforts based on previously disclosed unsubstituted 6,7-dihydro-4H-triazolo[4,5-c]pyridines, methyl substituted 5,6,7,8-tetrahydro[1,2,4]triazolo[4,3-a]pyrazines, and several other series lead to the identification of a series of 4-(R)-methyl-6,7-dihydro-4H-triazolo[4,5-c]pyridines that are selective P2X7 antagonists with potency at the rodent and human P2X7 ion channels. These novel P2X7 antagonists have suitable physicochemical properties, and several analogs have an excellent pharmacokinetic profile, good partitioning into the CNS and show robust in vivo target engagement after oral dosing. Improvements in metabolic stability led to the identification of JNJ-54175446 (14) as a candidate for clinical development. The drug discovery efforts and strategies that resulted in the identification of the clinical candidate are described herein.


Assuntos
Antagonistas do Receptor Purinérgico P2X/farmacologia , Piridinas/farmacologia , Receptores Purinérgicos P2X7/efeitos dos fármacos , Animais , Disponibilidade Biológica , Humanos , Antagonistas do Receptor Purinérgico P2X/farmacocinética
6.
Biochem Pharmacol ; 135: 1-11, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28214518

RESUMO

Maintaining the integrity of cellular membranes is critical to protecting metabolic activities and genetic information from the environment. Regulation of transport across membranes of essential chemicals, including water, nutrients, hormones and many drugs, is therefore key to cellular homeostasis and physiological processes. The two main transporter superfamilies are ATP-binding cassette (ABC) transporters that primarily function as efflux transporters, and the solute carrier (SLC) transporters. SLC transporters encompass 52 gene families with almost 400 different human transporter genes. Although long under-explored, SLC transporters are an emerging drug target class and the molecular target of several approved inhibitor drugs, such as selective serotonin reuptake inhibitors (SSRIs) for depression and sodium/glucose co-transporter (SGLT2) inhibitors for diabetes. Interestingly though, although loss-of-function mutations in numerous human SLC transporters are linked to Mendelian diseases, few reports of SLC transporter activators have appeared, and only inhibitors have been advanced to clinical studies. In this commentary, we discuss several strategies for potentiating SLC transporter function, from direct acting potentiators to modulators of transcription, translation or trafficking. We review the progress made in recent years toward the understanding of the structural and molecular basis of SLC transporter function and the pathways and mechanisms that regulate SLC expression, and describe the opportunities these new insights present for discovery of SLC transporter potentiators. Finally, we highlight the challenges associated with the various approaches and provide some thoughts on future directions that might facilitate the search for SLC potentiators with therapeutic potential.


Assuntos
Descoberta de Drogas/métodos , Proteínas de Membrana Transportadoras/metabolismo , Proteínas Carreadoras de Solutos/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Descoberta de Drogas/tendências , Humanos , Proteínas de Membrana Transportadoras/química , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Inibidores Seletivos de Recaptação de Serotonina/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Proteínas Carreadoras de Solutos/química
7.
PLoS One ; 12(1): e0170102, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28107393

RESUMO

Kv1.3 is a voltage-gated potassium channel expressed on T cells that plays an important role in T cell activation. Previous studies have shown that blocking Kv1.3 channels in human T cells during activation results in reduced calcium entry, cytokine production, and proliferation. The aim of the present study was to further explore the effects of Kv1.3 blockers on the response of different human T cell subsets under various stimulation conditions. Our studies show that, unlike the immune suppressor cyclosporine A, the inhibitory effect of Kv1.3 blockers was partial and stimulation strength dependent, with reduced inhibitory efficacy on T cells under strengthened anti-CD3/CD28 stimulations. T cell responses to allergens including house dust mites and ragweed were partially reduced by Kv1.3 blockers. The effect of Kv1.3 inhibition was dependent on T cell subsets, with stronger effects on CCR7- effector memory compared to CCR7+ central memory CD4 T cells. Calcium entry studies also revealed a population of CD4 T cells resistant to Kv1.3 blockade. Activation of CD4 T cells was accompanied with an increase in Kv1.3 currents but Kv1.3 transcripts were found to be reduced, suggesting a posttranscriptional mechanism in the regulation of Kv1.3 activities. In summary, Kv1.3 blockers inhibit T cell activation in a manner that is highly dependent on the T cell identity and stimulation strength, These findings suggest that Kv1.3 blockers inhibit T cells in a unique, conditional manner, further refining our understanding of the therapeutic potential of Kv1.3 blockers.


Assuntos
Canal de Potássio Kv1.3/antagonistas & inibidores , Ativação Linfocitária , Bloqueadores dos Canais de Potássio/farmacologia , Subpopulações de Linfócitos T , Linfócitos T/imunologia , Perfilação da Expressão Gênica , Humanos , Canal de Potássio Kv1.3/genética , Canal de Potássio Kv1.3/metabolismo , Técnicas de Patch-Clamp
8.
Mol Pharmacol ; 90(6): 766-774, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27754898

RESUMO

In the liver, citrate is a key metabolic intermediate involved in the regulation of glycolysis and lipid synthesis and reduced expression of the hepatic citrate SLC13A5 transporter has been shown to improve metabolic outcomes in various animal models. Although inhibition of hepatic extracellular citrate uptake through SLC13A5 has been suggested as a potential therapeutic approach for Type-2 diabetes and/or fatty liver disease, so far, only a few SLC13A5 inhibitors have been identified. Moreover, their mechanism of action still remains unclear, potentially limiting their utility for in vivo proof-of-concept studies. In this study, we characterized the pharmacology of the recently identified hydroxysuccinic acid SLC13A5 inhibitors, PF-06649298 and PF-06761281, using a combination of 14C-citrate uptake, a membrane potential assay and electrophysiology. In contrast to their previously proposed mechanism of action, our data suggest that both PF-06649298 and PF-06761281 are allosteric, state-dependent SLC13A5 inhibitors, with low-affinity substrate activity in the absence of citrate. As allosteric state-dependent modulators, the inhibitory potency of both compounds is highly dependent on the ambient citrate concentration and our detailed mechanism of action studies therefore, may be of value in interpreting the in vivo effects of these compounds.


Assuntos
Malatos/farmacologia , Fenilbutiratos/farmacologia , Piridinas/farmacologia , Succinatos/farmacologia , Simportadores/antagonistas & inibidores , Regulação Alostérica/efeitos dos fármacos , Radioisótopos de Carbono , Ácido Cítrico/metabolismo , Células HEK293 , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Malatos/química , Modelos Biológicos , Técnicas de Patch-Clamp , Fenilbutiratos/química , Piridinas/química , Especificidade por Substrato/efeitos dos fármacos , Succinatos/química , Simportadores/metabolismo
9.
Bioorg Med Chem Lett ; 26(19): 4781-4784, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27595421

RESUMO

The synthesis, SAR and preclinical characterization of a series of 6-chloro-N-(2-(4,4-difluoropiperidin-1-yl)-2-(2-(trifluoromethyl)pyrimidin-5-yl)ethyl)quinoline-5-carboxamide based P2X7 antagonists is described herein. The lead compounds are potent inhibitors in Ca(2+) flux and whole blood IL-1ß P2X7 release assays at both human and mouse isoforms. Compound 1e showed a robust reduction of IL-1ß release in a mouse ex vivo model with a 50mg/kg oral dose. Evaluation of compound 1e in the mouse SNI tactile allodynia, carrageenan-induced paw edema or CIA models resulted in no analgesic or anti-inflammatory effects.


Assuntos
Antagonistas do Receptor Purinérgico P2X/farmacologia , Quinolinas/farmacologia , Animais , Descoberta de Drogas , Humanos , Interleucina-1beta/metabolismo , Camundongos , Antagonistas do Receptor Purinérgico P2X/química , Quinolinas/química , Relação Estrutura-Atividade
10.
J Pharmacol Exp Ther ; 357(2): 394-414, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26989142

RESUMO

Members of the α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionic acid (AMPA) subtype of ionotropic glutamate receptors mediate the majority of fast synaptic transmission within the mammalian brain and spinal cord, representing attractive targets for therapeutic intervention. Here, we describe novel AMPA receptor modulators that require the presence of the accessory protein CACNG8, also known as transmembrane AMPA receptor regulatory protein γ8 (TARP-γ8). Using calcium flux, radioligand binding, and electrophysiological assays of wild-type and mutant forms of TARP-γ8, we demonstrate that these compounds possess a novel mechanism of action consistent with a partial disruption of the interaction between the TARP and the pore-forming subunit of the channel. One of the molecules, 5-[2-chloro-6-(trifluoromethoxy)phenyl]-1,3-dihydrobenzimidazol-2-one (JNJ-55511118), had excellent pharmacokinetic properties and achieved high receptor occupancy following oral administration. This molecule showed strong, dose-dependent inhibition of neurotransmission within the hippocampus, and a strong anticonvulsant effect. At high levels of receptor occupancy in rodent in vivo models, JNJ-55511118 showed a strong reduction in certain bands on electroencephalogram, transient hyperlocomotion, no motor impairment on rotarod, and a mild impairment in learning and memory. JNJ-55511118 is a novel tool for reversible AMPA receptor inhibition, particularly within the hippocampus, with potential therapeutic utility as an anticonvulsant or neuroprotectant. The existence of a molecule with this mechanism of action demonstrates the possibility of pharmacological targeting of accessory proteins, increasing the potential number of druggable targets.


Assuntos
Benzimidazóis/uso terapêutico , Canais de Cálcio/efeitos dos fármacos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Receptores de AMPA/efeitos dos fármacos , Animais , Canais de Cálcio/genética , Sinalização do Cálcio/efeitos dos fármacos , Desenho de Fármacos , Eletroencefalografia/efeitos dos fármacos , Células HEK293 , Humanos , Aprendizagem/efeitos dos fármacos , Memória/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Atividade Motora/efeitos dos fármacos , Mutação/genética , Neurônios/efeitos dos fármacos , Equilíbrio Postural/efeitos dos fármacos , Ratos Sprague-Dawley , Receptores de AMPA/genética
11.
J Biol Chem ; 289(33): 22704-22714, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24939846

RESUMO

Ion channels are an attractive class of drug targets, but progress in developing inhibitors for therapeutic use has been limited largely due to challenges in identifying subtype selective small molecules. Animal venoms provide an alternative source of ion channel modulators, and the venoms of several species, such as scorpions, spiders and snails, are known to be rich sources of ion channel modulating peptides. Importantly, these peptides often bind to hyper-variable extracellular loops, creating the potential for subtype selectivity rarely achieved with small molecules. We have engineered scorpion venom peptides and incorporated them in fusion proteins to generate highly potent and selective Kv1.3 inhibitors with long in vivo half-lives. Kv1.3 has been reported to play a role in human T cell activation, and therefore, these Kv1.3 inhibitor fusion proteins may have potential for the treatment of autoimmune diseases. Our results support an emerging approach to generating subtype selective therapeutic ion channel inhibitors.


Assuntos
Proteínas de Artrópodes/farmacologia , Canal de Potássio Kv1.3/antagonistas & inibidores , Ativação Linfocitária/efeitos dos fármacos , Peptídeos/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Engenharia de Proteínas , Venenos de Escorpião/farmacologia , Linfócitos T/metabolismo , Animais , Proteínas de Artrópodes/química , Proteínas de Artrópodes/genética , Células CHO , Cricetinae , Cricetulus , Sistemas de Liberação de Medicamentos , Meia-Vida , Humanos , Canal de Potássio Kv1.3/genética , Canal de Potássio Kv1.3/metabolismo , Peptídeos/química , Peptídeos/genética , Bloqueadores dos Canais de Potássio/química , Ratos , Venenos de Escorpião/química , Venenos de Escorpião/genética
12.
Proc Natl Acad Sci U S A ; 111(7): 2758-63, 2014 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-24497506

RESUMO

A cone snail venom peptide, µO§-conotoxin GVIIJ from Conus geographus, has a unique posttranslational modification, S-cysteinylated cysteine, which makes possible formation of a covalent tether of peptide to its target Na channels at a distinct ligand-binding site. µO§-conotoxin GVIIJ is a 35-aa peptide, with 7 cysteine residues; six of the cysteines form 3 disulfide cross-links, and one (Cys24) is S-cysteinylated. Due to limited availability of native GVIIJ, we primarily used a synthetic analog whose Cys24 was S-glutathionylated (abbreviated GVIIJSSG). The peptide-channel complex is stabilized by a disulfide tether between Cys24 of the peptide and Cys910 of rat (r) NaV1.2. A mutant channel of rNaV1.2 lacking a cysteine near the pore loop of domain II (C910L), was >10(3)-fold less sensitive to GVIIJSSG than was wild-type rNaV1.2. In contrast, although rNaV1.5 was >10(4)-fold less sensitive to GVIIJSSG than NaV1.2, an rNaV1.5 mutant with a cysteine in the homologous location, rNaV1.5[L869C], was >10(3)-fold more sensitive than wild-type rNaV1.5. The susceptibility of rNaV1.2 to GVIIJSSG was significantly altered by treating the channels with thiol-oxidizing or disulfide-reducing agents. Furthermore, coexpression of rNaVß2 or rNaVß4, but not that of rNaVß1 or rNaVß3, protected rNaV1.1 to -1.7 (excluding NaV1.5) against block by GVIIJSSG. Thus, GVIIJ-related peptides may serve as probes for both the redox state of extracellular cysteines and for assessing which NaVß- and NaVα-subunits are present in native neurons.


Assuntos
Conotoxinas/toxicidade , Dissulfetos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.2/metabolismo , Neurônios/metabolismo , Bloqueadores do Canal de Sódio Disparado por Voltagem/toxicidade , Sequência de Aminoácidos , Animais , Sequência de Bases , Cromatografia Líquida de Alta Pressão , Conotoxinas/genética , Conotoxinas/metabolismo , Cisteína/metabolismo , Primers do DNA/genética , DNA Complementar/genética , Dados de Sequência Molecular , Oócitos/metabolismo , Técnicas de Patch-Clamp , Ratos , Análise de Sequência de DNA , Espectrometria de Massas em Tandem , Bloqueadores do Canal de Sódio Disparado por Voltagem/metabolismo
13.
Curr Protoc Pharmacol ; 64: 11.1.1-17, 2014 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-26344208

RESUMO

This unit provides an overview of the principal electrophysiological techniques commonly used for the study of ionic currents and the ion channels that mediate them. These techniques include electroencephalograms (EEGs), electrocardiograms (ECGs), single- and multiunit extracellular recording, multielectrode arrays, transepithelial recording, impedance measurements, and current-clamp, voltage-clamp, patch-clamp, and lipid bilayer recording. The unit also discusses recent advances in high-throughput, automated electrophysiological techniques for drug discovery and the use of stem cells as a tissue source.


Assuntos
Eletrodiagnóstico/métodos , Animais , Descoberta de Drogas/métodos , Eletrocardiografia/métodos , Eletrocardiografia/normas , Eletrodiagnóstico/normas , Eletroencefalografia/métodos , Eletroencefalografia/normas , Fenômenos Eletrofisiológicos , Humanos , Técnicas de Patch-Clamp/métodos , Técnicas de Patch-Clamp/normas
14.
Br J Pharmacol ; 170(3): 624-40, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23889535

RESUMO

BACKGROUND AND PURPOSE: An increasing body of evidence suggests that the purinergic receptor P2X, ligand-gated ion channel, 7 (P2X7) in the CNS may play a key role in neuropsychiatry, neurodegeneration and chronic pain. In this study, we characterized JNJ-47965567, a centrally permeable, high-affinity, selective P2X7 antagonist. EXPERIMENTAL APPROACH: We have used a combination of in vitro assays (calcium flux, radioligand binding, electrophysiology, IL-1ß release) in both recombinant and native systems. Target engagement of JNJ-47965567 was demonstrated by ex vivo receptor binding autoradiography and in vivo blockade of Bz-ATP induced IL-1ß release in the rat brain. Finally, the efficacy of JNJ-47965567 was tested in standard models of depression, mania and neuropathic pain. KEY RESULTS: JNJ-47965567 is potent high affinity (pKi 7.9 ± 0.07), selective human P2X7 antagonist, with no significant observed speciation. In native systems, the potency of the compound to attenuate IL-1ß release was 6.7 ± 0.07 (human blood), 7.5 ± 0.07 (human monocytes) and 7.1 ± 0.1 (rat microglia). JNJ-47965567 exhibited target engagement in rat brain, with a brain EC50 of 78 ± 19 ng·mL(-1) (P2X7 receptor autoradiography) and functional block of Bz-ATP induced IL-1ß release. JNJ-47965567 (30 mg·kg(-1) ) attenuated amphetamine-induced hyperactivity and exhibited modest, yet significant efficacy in the rat model of neuropathic pain. No efficacy was observed in forced swim test. CONCLUSION AND IMPLICATIONS: JNJ-47965567 is centrally permeable, high affinity P2X7 antagonist that can be used to probe the role of central P2X7 in rodent models of CNS pathophysiology.


Assuntos
Encéfalo/efeitos dos fármacos , Niacinamida/análogos & derivados , Piperazinas/farmacologia , Antagonistas do Receptor Purinérgico P2X/farmacologia , Receptores Purinérgicos P2X7/efeitos dos fármacos , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Analgésicos/metabolismo , Analgésicos/farmacologia , Animais , Antidepressivos/farmacologia , Antimaníacos/farmacologia , Comportamento Animal/efeitos dos fármacos , Ligação Competitiva , Transtorno Bipolar/metabolismo , Transtorno Bipolar/prevenção & controle , Transtorno Bipolar/psicologia , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Permeabilidade Capilar , Linhagem Celular , Depressão/metabolismo , Depressão/prevenção & controle , Depressão/psicologia , Modelos Animais de Doenças , Cães , Relação Dose-Resposta a Droga , Humanos , Interleucina-1beta/metabolismo , Macaca , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neuralgia/metabolismo , Neuralgia/prevenção & controle , Neuralgia/psicologia , Niacinamida/metabolismo , Niacinamida/farmacologia , Piperazinas/metabolismo , Ligação Proteica , Antagonistas do Receptor Purinérgico P2X/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P2X7/metabolismo , Fatores de Tempo , Transfecção
15.
J Biol Chem ; 288(31): 22707-20, 2013 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-23760503

RESUMO

Voltage-gated sodium channels (VGSCs) are essential to the normal function of the vertebrate nervous system. Aberrant function of VGSCs underlies a variety of disorders, including epilepsy, arrhythmia, and pain. A large number of animal toxins target these ion channels and may have significant therapeutic potential. Most of these toxins, however, have not been characterized in detail. Here, by combining patch clamp electrophysiology and radioligand binding studies with peptide mutagenesis, NMR structure determination, and molecular modeling, we have revealed key molecular determinants of the interaction between the tarantula toxin huwentoxin-IV and two VGSC isoforms, Nav1.7 and Nav1.2. Nine huwentoxin-IV residues (F6A, P11A, D14A, L22A, S25A, W30A, K32A, Y33A, and I35A) were important for block of Nav1.7 and Nav1.2. Importantly, molecular dynamics simulations and NMR studies indicated that folding was normal for several key mutants, suggesting that these amino acids probably make specific interactions with sodium channel residues. Additionally, we identified several amino acids (F6A, K18A, R26A, and K27A) that are involved in isoform-specific VGSC interactions. Our structural and functional data were used to model the docking of huwentoxin-IV into the domain II voltage sensor of Nav1.7. The model predicts that a hydrophobic patch composed of Trp-30 and Phe-6, along with the basic Lys-32 residue, docks into a groove formed by the Nav1.7 S1-S2 and S3-S4 loops. These results provide new insight into the structural and molecular basis of sodium channel block by huwentoxin-IV and may provide a basis for the rational design of toxin-based peptides with improved VGSC potency and/or selectivity.


Assuntos
Ativação do Canal Iônico , Bloqueadores dos Canais de Sódio/farmacologia , Venenos de Aranha/química , Sequência de Aminoácidos , Células HEK293 , Humanos , Espectroscopia de Ressonância Magnética , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Ensaio Radioligante , Homologia de Sequência de Aminoácidos , Venenos de Aranha/farmacologia , Relação Estrutura-Atividade
16.
ACS Med Chem Lett ; 4(4): 419-22, 2013 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-24900687

RESUMO

The synthesis and preclinical characterization of two novel, brain penetrating P2X7 compounds will be described. Both compounds are shown to be high potency P2X7 antagonists in human, rat, and mouse cell lines and both were shown to have high brain concentrations and robust receptor occupancy in rat. Compound 7 is of particular interest as a probe compound for the preclinical assessment of P2X7 blockade in animal models of neuro-inflammation.

17.
Bioorg Med Chem Lett ; 21(18): 5197-201, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21824780

RESUMO

The discovery of a series of novel, potent, and selective blockers of the cyclic nucleotide-modulated channel HCN1 is disclosed. Here we report an SAR study around a series of selective blockers of the HCN1 channel. Utilization of a high-throughput VIPR assay led to the identification of a novel series of 2,2-disubstituted indane derivatives, which had moderate selectivity and potency at HCN1. Optimization of this hit led to the identification of the potent, 1,1-disubstituted cyclohexane HCN1 blocker, 2-ethoxy-N-((1-(4-isopropylpiperazin-1-yl)cyclohexyl)methyl)benzamide. The work leading to the discovery of this compound is described herein.


Assuntos
Canais de Cátion Regulados por Nucleotídeos Cíclicos/antagonistas & inibidores , Descoberta de Drogas , Indanos/farmacologia , Animais , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização , Indanos/síntese química , Indanos/química , Camundongos , Estrutura Molecular , Canais de Potássio/metabolismo , Estereoisomerismo , Relação Estrutura-Atividade
18.
Eur J Pharmacol ; 663(1-3): 40-50, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21575625

RESUMO

As an integrator of multiple nociceptive and/or inflammatory stimuli, TRPV1 is an attractive therapeutic target for the treatment of various painful disorders. Several TRPV1 antagonists have been advanced into clinical trials and the initial observations suggest that TRPV1 antagonism may be associated with mild hyperthermia and thermal insensitivity in man. However, no clinical efficacy studies have been described to date, making an assessment of risk:benefit impossible. Furthermore, it is not clear whether these early observations are representative of all TRPV1 antagonists and whether additional clinical studies with novel TRPV1 antagonists are required in order to understand optimal compound characteristics. In the present study we describe 2-(2,6-dichloro-benzyl)-thiazolo[5,4-d]pyrimidin-7-yl]-(4-trifluoromethyl-phenyl)-amine (JNJ-39729309) as a novel, TRPV1 antagonist. JNJ-39729209 displaced tritiated resiniferotoxin binding to TRPV1 and prevented TRPV1 activation by capsaicin, protons and heat. In-vivo, JNJ-39729209 blocked capsaicin-induced hypotension, induced a mild hyperthermia and inhibited capsaicin-induced hypothermia in a dose dependent manner. JNJ-39729209 showed significant efficacy against carrageenan- and CFA-evoked thermal hyperalgesia and exhibited significant anti-tussive activity in a guinea-pig model of capsaicin-induced cough. In pharmacokinetic studies, JNJ-39729209 was found to have low clearance, a moderate volume of distribution, good oral bioavailability and was brain penetrant. On the basis of these findings, JNJ-39729209 represents a structurally novel TRPV1 antagonist with potential for clinical development. The advancement of JNJ-39729209 into human clinical trials could be useful in further understanding the analgesic potential of TRPV1 antagonists.


Assuntos
Pirimidinas/farmacologia , Canais de Cátion TRPV/antagonistas & inibidores , Tiazóis/farmacologia , Animais , Temperatura Corporal/efeitos dos fármacos , Linhagem Celular , Ensaios Clínicos como Assunto , Tosse/tratamento farmacológico , Cães , Feminino , Cobaias , Humanos , Hiperalgesia/tratamento farmacológico , Hipotensão/tratamento farmacológico , Masculino , Camundongos , Pirimidinas/farmacocinética , Pirimidinas/uso terapêutico , Ratos , Tiazóis/farmacocinética , Tiazóis/uso terapêutico
19.
Curr Pharm Biotechnol ; 12(10): 1698-706, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21466444

RESUMO

P2X7 is an ATP-gated non-selective cation channel expressed primarily on cells of hematopoietic origin, such as macrophages and microglia. Since the initial cloning of this channel, enormous progress has been made in the understanding of the physiology, pharmacology and therapeutic utility of P2X7. This article attempts to review the biology of P2X7 with a focus on the complex pharmacology of this channel. Finally, the authors discuss the role of P2X7 as an analgesic drug target and raise some of the challenges and issues that face the P2X7 research community.


Assuntos
Dor/metabolismo , Receptores Purinérgicos P2X7/fisiologia , Analgésicos/uso terapêutico , Animais , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Dor/tratamento farmacológico
20.
ACS Med Chem Lett ; 2(6): 481-4, 2011 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-24900334

RESUMO

A series of N-pyridyl benzamide KCNQ2/Q3 potassium channel openers were identified and found to be active in animal models of epilepsy and pain. The best compound 12 [ICA-027243, N-(6-chloro-pyridin-3-yl)-3,4-difluoro-benzamide] has an EC50 of 0.38 µM and is selective for KCNQ2/Q3 channels. This compound was active in several rodent models of epilepsy and pain but upon repeated dosing had a number of unacceptable toxicities that prevented further development. On the basis of the structure-activity relationships developed around 12, a second compound, 51, [N-(2-chloro-pyrimidin-5-yl)-3,4-difluoro-benzamide, ICA-069673], was prepared and advanced into a phase 1 clinical study. Herein, we describe the structure-activity relationships that led to the identification of compound 12 and to the corresponding pyrimidine 51.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA