Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Biomolecules ; 13(6)2023 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-37371516

RESUMO

Opioid analgesics such as morphine and fentanyl induce mu-opioid receptor (MOR)-mediated hyperactivity in mice. Herein, we show that morphine, fentanyl, SR-17018, and oliceridine have submaximal intrinsic efficacy in the mouse striatum using 35S-GTPγS binding assays. While all of the agonists act as partial agonists for stimulating G protein coupling in striatum, morphine, fentanyl, and oliceridine are fully efficacious in stimulating locomotor activity; meanwhile, the noncompetitive biased agonists SR-17018 and SR-15099 produce submaximal hyperactivity. Moreover, the combination of SR-17018 and morphine attenuates hyperactivity while antinociceptive efficacy is increased. The combination of oliceridine with morphine increases hyperactivity, which is maintained over time. These findings provide evidence that noncompetitive agonists at MOR can be used to suppress morphine-induced hyperactivity while enhancing antinociceptive efficacy; moreover, they demonstrate that intrinsic efficacy measured at the receptor level is not directly proportional to drug efficacy in the locomotor activity assay.


Assuntos
Morfina , Compostos de Espiro , Camundongos , Animais , Morfina/farmacologia , Analgésicos Opioides/farmacologia , Fentanila/farmacologia
2.
SLAS Discov ; 27(1): 8-19, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35058179

RESUMO

The severe acute respiratory syndrome coronavirus 2 responsible for COVID-19 remains a persistent threat to mankind, especially for the immunocompromised and elderly for which the vaccine may have limited effectiveness. Entry of SARS-CoV-2 requires a high affinity interaction of the viral spike protein with the cellular receptor angiotensin-converting enzyme 2. Novel mutations on the spike protein correlate with the high transmissibility of new variants of SARS-CoV-2, highlighting the need for small molecule inhibitors of virus entry into target cells. We report the identification of such inhibitors through a robust high-throughput screen testing 15,000 small molecules from unique libraries. Several leads were validated in a suite of mechanistic assays, including whole cell SARS-CoV-2 infectivity assays. The main lead compound, calpeptin, was further characterized using SARS-CoV-1 and the novel SARS-CoV-2 variant entry assays, SARS-CoV-2 protease assays and molecular docking. This study reveals calpeptin as a potent and specific inhibitor of SARS-CoV-2 and some variants.


Assuntos
Antivirais/farmacologia , Tratamento Farmacológico da COVID-19 , Inibidores de Cisteína Proteinase/farmacologia , Dipeptídeos/farmacologia , Ligação Viral/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Enzima de Conversão de Angiotensina 2/metabolismo , Animais , Catepsina L/antagonistas & inibidores , Linhagem Celular , Chlorocebus aethiops , Avaliação Pré-Clínica de Medicamentos , Reposicionamento de Medicamentos , Células HEK293 , Humanos , Simulação de Acoplamento Molecular , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/crescimento & desenvolvimento , Serina Endopeptidases/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo , Células Vero
3.
Proc Natl Acad Sci U S A ; 118(48)2021 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-34819362

RESUMO

The ability of a ligand to preferentially promote engagement of one signaling pathway over another downstream of GPCR activation has been referred to as signaling bias, functional selectivity, and biased agonism. The presentation of ligand bias reflects selectivity between active states of the receptor, which may result in the display of preferential engagement with one signaling pathway over another. In this study, we provide evidence that the G protein-biased mu opioid receptor (MOR) agonists SR-17018 and SR-14968 stabilize the MOR in a wash-resistant yet antagonist-reversible G protein-signaling state. Furthermore, we demonstrate that these structurally related biased agonists are noncompetitive for radiolabeled MOR antagonist binding, and while they stimulate G protein signaling in mouse brains, partial agonists of this class do not compete with full agonist activation. Importantly, opioid antagonists can readily reverse their effects in vivo. Given that chronic treatment with SR-17018 does not lead to tolerance in several mouse pain models, this feature may be desirable for the development of long-lasting opioid analgesics that remain sensitive to antagonist reversal of respiratory suppression.


Assuntos
Receptores Acoplados a Proteínas G/metabolismo , Receptores Opioides mu/metabolismo , Transdução de Sinais/efeitos dos fármacos , Analgésicos Opioides/farmacologia , Animais , Benzimidazóis/farmacologia , Proteínas de Ligação ao GTP/metabolismo , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Antagonistas de Entorpecentes/farmacologia , Piperidinas/farmacologia , Receptores Acoplados a Proteínas G/fisiologia , Receptores Opioides mu/agonistas , Receptores Opioides mu/fisiologia , Transdução de Sinais/fisiologia , beta-Arrestina 2/metabolismo
4.
Neuropharmacology ; 185: 108439, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33345829

RESUMO

The mu opioid receptor-selective agonist, SR-17018, preferentially activates GTPγS binding over ßarrestin2 recruitment in cellular assays, thereby demonstrating signaling bias. In mice, SR-17018 stimulates GTPγS binding in brainstem and produces antinociception with potencies similar to morphine. However, it produces much less respiratory suppression and mice do not develop antinociceptive tolerance in the hot plate assay upon repeated dosing. Herein we evaluate the effects of acute and repeated dosing of SR-17018, oxycodone and morphine in additional models of pain-related behaviors. In the mouse warm water tail immersion assay, an assessment of spinal reflex to thermal nociception, repeated administration of SR-17018 produces tolerance as does morphine and oxycodone. SR-17018 retains efficacy in a formalin-induced inflammatory pain model upon repeated dosing, while oxycodone does not. In a chemotherapeutic-induced neuropathy pain model SR-17018 is more potent and efficacious than morphine or oxycodone, moreover, this efficacy is retained upon repeated dosing of SR-17018. These findings demonstrate that, with the exception of the tail flick test, SR-17018 retains efficacy upon chronic treatment across several pain models.


Assuntos
Analgésicos Opioides/administração & dosagem , Benzimidazóis/administração & dosagem , Morfina/administração & dosagem , Neuralgia/tratamento farmacológico , Oxicodona/administração & dosagem , Piperidinas/administração & dosagem , Receptores Opioides mu/agonistas , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Tolerância a Medicamentos , Feminino , Bombas de Infusão Implantáveis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Neuralgia/patologia , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Resultado do Tratamento
5.
Neuropsychopharmacology ; 45(2): 416-425, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31443104

RESUMO

It has been demonstrated that opioid agonists that preferentially act at µ-opioid receptors to activate G protein signaling over ßarrestin2 recruitment produce antinociception with less respiratory suppression. However, most of the adverse effects associated with opioid therapeutics are realized after extended dosing. Therefore, we tested the onset of tolerance and dependence, and assessed for neurochemical changes associated with prolonged treatment with the biased agonist SR-17018. When chronically administered to mice, SR-17018 does not lead to hot plate antinociceptive tolerance, receptor desensitization in periaqueductal gray, nor a super-sensitization of adenylyl cyclase in the striatum, which are hallmarks of opioid neuronal adaptations that are seen with morphine. Interestingly, substitution with SR-17018 in morphine-tolerant mice restores morphine potency and efficacy, whereas the onset of opioid withdrawal is prevented. This is in contrast to buprenorphine, which can suppress withdrawal, but produces and maintains morphine antinociceptive tolerance. Biased agonists of this nature may therefore be useful for the treatment of opioid dependence while restoring opioid antinociceptive sensitivity.


Assuntos
Analgésicos Opioides/metabolismo , Tolerância a Medicamentos/fisiologia , Dependência de Morfina/metabolismo , Morfina/metabolismo , Receptores Opioides mu/metabolismo , Síndrome de Abstinência a Substâncias/metabolismo , Analgésicos Opioides/administração & dosagem , Animais , Relação Dose-Resposta a Droga , Feminino , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Bombas de Infusão Implantáveis , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/administração & dosagem , Oxicodona/administração & dosagem , Oxicodona/metabolismo , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Receptores Opioides mu/agonistas , Síndrome de Abstinência a Substâncias/prevenção & controle
6.
J Med Chem ; 61(19): 8895-8907, 2018 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-30199635

RESUMO

While mu opioid receptor (MOR) agonists are especially effective as broad-spectrum pain relievers, it has been exceptionally difficult to achieve a clear separation of analgesia from many problematic side effects. Recently, many groups have sought MOR agonists that induce minimal ßarrestin-mediated signaling because MOR agonist-treated ßarrestin2 knockout mice were found to display enhanced antinociceptive effects with significantly less respiratory depression and tachyphylaxis. Substantial data now exists to support the premise that G protein signaling biased MOR agonists can be effective analgesic agents. We recently showed that, within a chemical series, the degree of bias correlates linearly with the magnitude of the respiratory safety index. Herein we describe the synthesis and optimization of piperidine benzimidazolone MOR agonists that together display a wide range of bias (G/ßarr2). We identify structural features affecting potency and maximizing bias and show that many compounds have desirable properties, such as long half-lives and high brain penetration.


Assuntos
Analgésicos Opioides/farmacologia , Barreira Hematoencefálica/metabolismo , Descoberta de Drogas/normas , Proteínas de Ligação ao GTP/metabolismo , Microssomos Hepáticos/metabolismo , Receptores Opioides mu/agonistas , Analgésicos Opioides/química , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microssomos Hepáticos/efeitos dos fármacos , Conformação Proteica , Relação Estrutura-Atividade , beta-Arrestinas/metabolismo
7.
Cell ; 171(5): 1165-1175.e13, 2017 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-29149605

RESUMO

Biased agonism has been proposed as a means to separate desirable and adverse drug responses downstream of G protein-coupled receptor (GPCR) targets. Herein, we describe structural features of a series of mu-opioid-receptor (MOR)-selective agonists that preferentially activate receptors to couple to G proteins or to recruit ßarrestin proteins. By comparing relative bias for MOR-mediated signaling in each pathway, we demonstrate a strong correlation between the respiratory suppression/antinociception therapeutic window in a series of compounds spanning a wide range of signaling bias. We find that ßarrestin-biased compounds, such as fentanyl, are more likely to induce respiratory suppression at weak analgesic doses, while G protein signaling bias broadens the therapeutic window, allowing for antinociception in the absence of respiratory suppression.


Assuntos
Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/efeitos adversos , Receptores Opioides mu/agonistas , Animais , Fentanila/administração & dosagem , Proteínas de Ligação ao GTP/metabolismo , Camundongos , Morfina/administração & dosagem , Receptores Opioides mu/química , Sistema Respiratório/efeitos dos fármacos , Transdução de Sinais , beta-Arrestinas/metabolismo
8.
J Food Prot ; 79(7): 1259-65, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27357048

RESUMO

Contamination by and persistence of pathogenic bacteria in ready-to-eat produce have emerged as significant food safety and public health concerns. Viable produceborne pathogens cope with several stresses (e.g., temperature fluctuations and lowtemperature storage) during production and storage of the commodities. In this study, we investigated the impact of transient cold shock on Escherichia coli O157:H7 (EcO157) cells in a produce matrix (romaine lettuce leaf lysate). EcO157 cells were exposed to 25°C for 1 h, 4°C for 1 h, and 4°C for 10 min in lettuce lysate. The expression of selected genes coding for virulence, stress response, and heat and cold shock proteins was quantified by real-time quantitative reverse transcription PCR assay. Treated EcO157 cells adhered to MAC-T mammalian cells were enumerated by in vitro bioassay. Expression of the Shiga toxin 1 gene (stx1a) was upregulated significantly (P < 0.05) upon cold shock treatments, but virulence genes related to EcO157 attachment (eaeA, lpfA, and hcpA) were down-regulated. Two key members of the cold shock regulon, cold shock protein (cspA) and gyrA, were significantly induced (P < 0.05) at the refrigeration temperature (4°C). Significant upregulation of an SOS response gene, recA, was also observed. E. coli heat shock regulon member grpE was induced, but a universal stress protein (uspA) was downregulated at the refrigeration temperatures in lettuce lysate. The adhesion assay revealed a temperature-dependent reduction in the attachment of cold-shocked EcO157 cells. The results of the current study indicate a reduction in the attachment of cold-shocked EcO157 to epithelial cells and higher levels of Shiga toxin gene expression at the molecular level.


Assuntos
Escherichia coli O157 , Lactuca/microbiologia , Animais , Contagem de Colônia Microbiana , Microbiologia de Alimentos , Folhas de Planta/microbiologia , Toxina Shiga I , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA