Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Pharmacol ; 98(4): 462-474, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32958572

RESUMO

Opioid receptors (ORs) convert extracellular messages to signaling events by coupling to the heterotrimeric G proteins, Gα•ßγ Classic pharmacological methods, such as [35S]GTPγS binding and inhibition of cyclic AMP production, allow for general opioid characterization, but they are subject to the varying endogenous Gα proteins in a given cell type. Bioluminescence resonance energy transfer (BRET) technology offers new insight by allowing the direct observation of Gα subunit-specific effects on opioid pharmacology. Using a Venus-tagged Gßγ and nanoluciferase-tagged truncated G protein receptor kinase 3, an increase in BRET signal correlated with OR activation mediated by a specific Gα protein. The magnitude of the BRET signal was normalized to the maximum response obtained with 10 µM 2-(3,4-dichlorophenyl)-N-methyl-N-[(1R,2R)-2-pyrrolidin-1-ylcyclohexyl]acetamide (U50,488) for the kappa OR (KOR). Opioids reached equilibrium with the KOR, and concentration-response curves were generated. Although the full agonists U50,488, salvinorin A, nalfurafine, and dynorphin peptides were equally efficacious regardless of the Gα subunit present, the concentration-response curves were leftward shifted when the KOR was signaling through Gαz compared with other Gαi/o subunits. In contrast, the Gα subunit distinctly affected both the efficacy and potency of partial kappa agonists, such as the benzomorphans, and the classic mu opioid antagonists, naloxone, naltrexone, and nalmefene. For example, (-)pentazocine had EC50 values of 7.3 and 110 nM and maximal stimulation values of 79% and 35% when the KOR signaled through Gαz and Gαi1, respectively. Together, these observations suggest KOR pharmacology varies based on the specific Gα subunit coupled to the KOR. SIGNIFICANCE STATEMENT: Opioid receptors couple to various heterotrimeric Gαßγ proteins to convert extracellular cues to precise intracellular events. This paper focuses on how the various inhibitory Gα subunits influence the pharmacology of full and partial agonists at the kappa opioid receptor. Using a bioluminescent assay, the efficacy and potency of kappa opioids was determined. Opioid signaling was more potent through Gαz compared with other Gα proteins. These observations suggest that Gαz may impact opioid pharmacology and cellular physiology more than previously thought.


Assuntos
Analgésicos Opioides/farmacologia , Técnicas de Transferência de Energia por Ressonância de Bioluminescência/métodos , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Receptores Opioides kappa/metabolismo , (trans)-Isômero de 3,4-dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclo-hexil)-benzenoacetamida/farmacologia , Diterpenos Clerodânicos/farmacologia , Dinorfinas/farmacologia , Células HEK293 , Humanos , Morfinanos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Compostos de Espiro/farmacologia
2.
Int J Mol Sci ; 22(1)2020 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-33374986

RESUMO

In previous work we evaluated an opioid glycopeptide with mixed µ/δ-opioid receptor agonism that was a congener of leu-enkephalin, MMP-2200. The glycopeptide analogue showed penetration of the blood-brain barrier (BBB) after systemic administration to rats, as well as profound central effects in models of Parkinson's disease (PD) and levodopa (L-DOPA)-induced dyskinesia (LID). In the present study, we tested the glycopeptide BBI-11008 with selective δ-opioid receptor agonism, an analogue of deltorphin, a peptide secreted from the skin of frogs (genus Phyllomedusa). We tested BBI-11008 for BBB-penetration after intraperitoneal (i.p.) injection and evaluated effects in LID rats. BBI-11008 (10 mg/kg) demonstrated good CNS-penetrance as shown by microdialysis and mass spectrometric analysis, with peak concentration levels of 150 pM in the striatum. While BBI-11008 at both 10 and 20 mg/kg produced no effect on levodopa-induced limb, axial and oral (LAO) abnormal involuntary movements (AIMs), it reduced the levodopa-induced locomotor AIMs by 50% after systemic injection. The N-methyl-D-aspartate receptor antagonist MK-801 reduced levodopa-induced LAO AIMs, but worsened PD symptoms in this model. Co-administration of MMP-2200 had been shown prior to block the MK-801-induced pro-Parkinsonian activity. Interestingly, BBI-11008 was not able to block the pro-Parkinsonian effect of MK-801 in the LID model, further indicating that a balance of mu- and delta-opioid agonism is required for this modulation. In summary, this study illustrates another example of meaningful BBB-penetration of a glycopeptide analogue of a peptide to achieve a central behavioral effect, providing additional evidence for the glycosylation technique as a method to harness therapeutic potential of peptides.


Assuntos
Modelos Animais de Doenças , Discinesia Induzida por Medicamentos/fisiopatologia , Glicopeptídeos/farmacologia , Atividade Motora/efeitos dos fármacos , Doença de Parkinson Secundária/fisiopatologia , Receptores Opioides delta/agonistas , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/farmacocinética , Analgésicos Opioides/farmacologia , Animais , Corpo Estriado/metabolismo , Maleato de Dizocilpina/farmacologia , Discinesia Induzida por Medicamentos/metabolismo , Glicopeptídeos/administração & dosagem , Glicopeptídeos/farmacocinética , Levodopa , Masculino , Atividade Motora/fisiologia , Fármacos Neuroprotetores/farmacologia , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/metabolismo , Ratos Sprague-Dawley , Receptores Opioides delta/metabolismo
3.
J Pharmacol Exp Ther ; 367(2): 267-281, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30108159

RESUMO

A combination of buprenorphine (BUP) and samidorphan (SAM) at a 1:1 (mg/mg) fixed-ratio dose is being investigated as an adjunctive treatment of major depressive disorder (BUP/SAM, ALKS 5461). Both [3H]BUP and [3H]SAM bound to the µ-, κ-, and δ-opioid receptors (MOR, KOR, and DOR, respectively) with Kd values of 3 nM or less. [3H]BUP dissociated from the MOR more slowly than [3H]SAM did. In the [35S]GTPγS assay, BUP was a partial agonist at the MOR, KOR, and DOR. SAM was an antagonist at the MOR and a partial agonist at the KOR and DOR. The pharmacology of the combination of SAM and BUP was characterized at ratios like the molar ratios of both compounds at steady state in humans. In all assessments, SAM reduced the efficacy of BUP at the MOR without altering its potency. At the KOR, SAM had no significant effect on the activity of BUP. In bioluminescent resonance energy transfer assays, SAM, naltrexone, and naloxone were partial agonists when the MOR was coupled to the Gα oB and Gα z, and were antagonists when coupled to Gα i At the KOR, SAM was a partial agonist activating Gα oA and Gα oB and a full agonist in stimulating Gα z SAM inhibited BUP's recruitment of ß-arrestin to the MOR, suggesting an attenuation of BUP's efficacy in activating G proteins correlated with an inhibition of ß-arrestin recruitment. The collective data suggest that SAM attenuates the efficacy of BUP under all conditions tested at the MOR and DOR but had little effect on BUP activity at the KOR.


Assuntos
Buprenorfina/farmacologia , Transtorno Depressivo Maior/tratamento farmacológico , Naltrexona/análogos & derivados , Animais , Células CHO , Linhagem Celular , Cricetulus , Combinação de Medicamentos , Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Naloxona/farmacologia , Naltrexona/farmacologia , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , beta-Arrestinas/metabolismo
4.
Brain Behav Immun ; 67: 152-162, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28844812

RESUMO

Patients receiving the cytokine immunotherapy, interferon-alpha (IFN-α) frequently present with neuropsychiatric consequences and cognitive impairments. Patients (25-80%) report symptoms of depression, including, anhedonia, irritability, fatigue and impaired motivation. Our lab has previously demonstrated treatment (170,000IU/kg sc, 3 times per week for 4weeks) of the pro-inflammatory cytokine, IFN-α, induced a depressive phenotype in rats in the forced swim test (FST). Here, we examine the biological mechanisms underlying behavioral changes induced by IFN-α, which may be reflective of mechanisms underlying inflammation associated depression. We also investigate the potential of 3-carboxamido seco-nalmefene (3CS-nalmefene), a novel opioid modulator (antagonist at mu and partial agonist at kappa and delta opioid receptors in vitro), to reverse IFN-α induced changes. In vitro radioligand receptor binding assays and the [35S] GTPγS were performed to determine the affinity of 3CS-nalmefene for the mu, kappa and delta opioid receptors. IFN-α treatment increased circulating and central markers of inflammation and hypothalamic-pituitaryadrenal (HPA) axis activity (IL-6, IL-1ß and corticosterone) while increasing immobility in the FST, impairing of object displacement learning in the object exploration task (OET), and decreasing neuronal proliferation and brain-derived neurotrophic factor (BDNF) in the hippocampus. Treatment with 3CS-nalmefene (0.3mg/kg/sc twice per day, 3 times per week for 4weeks) prevented IFN-α-induced immobility in the FST and impaired object displacement learning. In addition, 3CS-nalmefene prevented IFN-α-induced increases in inflammation and hyperactivity of the HPA-axis, the IFN-α-induced reduction in both neuronal proliferation and BDNF expression in the hippocampus. Overall, these preclinical data would support the hypothesis that opioid receptor modulation is a relevant target for treatment of depression.


Assuntos
Antidepressivos/administração & dosagem , Transtorno Depressivo/tratamento farmacológico , Naltrexona/análogos & derivados , Antagonistas de Entorpecentes/administração & dosagem , Receptores Opioides/agonistas , Animais , Ansiedade/induzido quimicamente , Ansiedade/tratamento farmacológico , Comportamento Animal/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Proliferação de Células/efeitos dos fármacos , Depressão/induzido quimicamente , Depressão/tratamento farmacológico , Transtorno Depressivo/induzido quimicamente , Modelos Animais de Doenças , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Interferon-alfa/administração & dosagem , Masculino , Naltrexona/administração & dosagem , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Ratos Wistar
5.
Behav Pharmacol ; 24(2): 144-52, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23412114

RESUMO

Inhibition of Gßγ-subunit signaling to phospholipase C ß3 has been shown to potentiate morphine-mediated antinociception while attenuating the development of tolerance and dependence in mice. The objective of this study was to determine the effect of Gßγ-subunit inhibition on antinociception and other pharmacological effects, such as respiratory depression, constipation, and hyperlocomotion, mediated by the µ-opioid receptor. The Gßγ-subunit inhibitor, gallein, was administered to C57BL/6J mice by intraperitoneal injection before morphine, and data were compared with mice treated with vehicle, morphine, or gallein alone. Morphine-induced antinociception was measured using the 55°C warm-water tail-withdrawal test. Pretreatment with gallein produced a dose-dependent potentiation of morphine-mediated antinociception, producing up to a 10-fold leftward shift in the morphine dose-response curve and extending the duration of antinociception induced by a single dose of morphine. Gallein pretreatment also prevented acute antinociceptive tolerance induced by morphine. In contrast, the dose-dependent respiratory depression and hyperlocomotion induced by morphine were not potentiated by gallein pretreatment. Similarly, gallein pretreatment did not potentiate morphine-conditioned place preference responses or morphine-induced constipation, as measured as a reduction in excreta. These results suggest that selectively inhibiting Gßγ-mediated signaling may selectively increase µ-opioid receptor-mediated antinociception without matching increases in adverse physiological effects.


Assuntos
Analgésicos Opioides/agonistas , Constipação Intestinal/prevenção & controle , Subunidades beta da Proteína de Ligação ao GTP/antagonistas & inibidores , Subunidades gama da Proteína de Ligação ao GTP/antagonistas & inibidores , Hipercinese/prevenção & controle , Morfina/agonistas , Insuficiência Respiratória/prevenção & controle , Xantenos/uso terapêutico , Analgésicos Opioides/administração & dosagem , Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/uso terapêutico , Animais , Comportamento Animal/efeitos dos fármacos , Constipação Intestinal/induzido quimicamente , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Tolerância a Medicamentos , Hipercinese/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Morfina/administração & dosagem , Morfina/efeitos adversos , Morfina/uso terapêutico , Atividade Motora/efeitos dos fármacos , Medição da Dor , Receptores Opioides mu/agonistas , Insuficiência Respiratória/induzido quimicamente , Recompensa , Sistemas do Segundo Mensageiro/efeitos dos fármacos
6.
Bioorg Med Chem Lett ; 23(7): 2128-33, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23434225

RESUMO

Derivatives of the lead compound N-BPE-8-CAC (1) where each CH of the biphenyl group was individually replaced by N were prepared in hopes of identifying high affinity ligands with improved aqueous solubility. Compared to 1, binding affinities of the five possible pyridinyl derivatives for the µ opioid receptor were between threefold lower to fivefold higher with the Ki of the most potent compound being 0.064 nM. Docking of 8-CAC (2) into the unliganded binding site of the mouse µ opioid receptor (pdb: 4DKL) revealed that 8-CAC and ß-FNA (from 4DKL) make nearly identical interactions with the receptor. However, for 1 and the new pyridinyl derivatives 4-8, binding is not tolerated in the 8-CAC binding mode due to the steric constraints of the large N-substituents. Either an alternative binding mode or rearrangement of the protein to accommodate these modifications may account for their high binding affinity.


Assuntos
Compostos de Bifenilo/farmacologia , Ciclopropanos/farmacologia , Piridinas/farmacologia , Receptores Opioides/química , Animais , Compostos de Bifenilo/síntese química , Compostos de Bifenilo/química , Ciclopropanos/síntese química , Ciclopropanos/química , Relação Dose-Resposta a Droga , Humanos , Ligantes , Camundongos , Modelos Moleculares , Estrutura Molecular , Piridinas/síntese química , Piridinas/química , Receptores Opioides/metabolismo , Relação Estrutura-Atividade
7.
NeuroImmune Pharm Ther ; 2(2): 187-244, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37476290

RESUMO

The 27th Scientific Conference of the Society on Neuroimmune Pharmacology (SNIP) in New Delhi, India, on March 15-18, 2023 is a historic summit of experts from around the world. The four day conference provides insights into the latest and most advanced science in the intersecting areas of neuroscience, immunology, pharmacology, and its translational aspects, in particular, HIV and drug abuse. Abstracts are ordered in three major groups: (1) Symposium speakers (S1-S64), (2) Investigator Posters (I1-I18), and (3) Trainee Poster (T1-T28).

8.
Bioorg Med Chem Lett ; 22(24): 7340-4, 2012 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-23142613

RESUMO

N-[2-(4'-methoxy[1,1'-biphenyl]-4-yl)ethyl]-8-CAC (1) is a high affinity (K(i)=0.084 nM) ligand for the µ opioid receptor and served as the lead compound for this study. Analogues of 1 were made in hopes of identifying an SAR within a series of oxygenated (distal) phenyl derivatives. A number of new analogues were made having single-digit pM affinity for the µ receptor. The most potent was the 3',4'-methylenedioxy analogue 18 (K(i)=1.6 pM).


Assuntos
Ciclazocina/análogos & derivados , Oxigênio/química , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Ciclazocina/síntese química , Ciclazocina/química , Ciclazocina/farmacologia , Relação Dose-Resposta a Droga , Humanos , Ligantes , Estrutura Molecular , Relação Estrutura-Atividade
9.
Neuropharmacology ; 202: 108858, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34715121

RESUMO

Because of increased opioid misuse, there is a need to identify new targets for minimizing opioid tolerance, and physical and psychological dependence. Previous studies showed that fibroblast growth factor 21 (FGF21) decreased alcohol and sweet preference in mice. In this study, FGF21-transgenic (FGF21-Tg) mice, expressing high FGF21 serum levels, and wildtype (WT) C57BL/6J littermates were treated with morphine and saline to determine if differences exist in their physiological and behavioral responses to opioids. FGF21-Tg mice displayed reduced preference for morphine in the conditioned place preference assay compared to WT littermates. Similarly, FGF21-Tg mice had an attenuation of the magnitude and rate of acute morphine antinociceptive tolerance development, and acute and chronic morphine physical dependence, but exhibited no change in chronic morphine antinociceptive tolerance. The ED50 values for morphine-induced antinociception in the 55 °C hot plate and the 55 °C warm-water tail withdrawal assays were similar in both strains of mice. Likewise, FGF21-Tg and WT littermates had comparable responses to morphine-induced respiratory depression. Overall, FGF21-Tg mice had a decrease in the development of acute analgesic tolerance, and the development of physical dependence, and morphine preference. FGF21 and its receptor have therapeutic potential for reducing opioid withdrawal symptoms and craving, and augmenting opioid therapeutics for acute pain patients to minimize tolerance development.


Assuntos
Tolerância a Medicamentos/genética , Fatores de Crescimento de Fibroblastos/sangue , Fatores de Crescimento de Fibroblastos/fisiologia , Dependência de Morfina/genética , Morfina/efeitos adversos , Nociceptividade/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Insuficiência Respiratória/induzido quimicamente , Insuficiência Respiratória/genética , Síndrome de Abstinência a Substâncias/genética , Síndrome de Abstinência a Substâncias/terapia
10.
J Pharmacol Exp Ther ; 336(3): 767-78, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21118955

RESUMO

We have previously reported the chemistry and antinociceptive properties of a series of glycosylated enkephalin analogs (glycopeptides) exhibiting approximately equal affinity and efficacy at δ opioid receptors (DORs) and µ opioid receptors (MORs). More detailed pharmacology of the lead glycopeptide MMP-2200 [H2N-Tyr-D-Thr-Gly-Phe-Leu-Ser-(O-ß-D-lactose)-CONH2] is presented. MMP-2200 produced dose-related antinociception in the 55°C tail-flick assay after various routes of administration. The antinociceptive effects of MMP-2200 were blocked by pretreatment with the general opioid antagonist naloxone and partially blocked by the MOR-selective antagonist ß-funaltrexamine and the DOR-selective antagonist naltrindole. The κ opioid receptor antagonist nor-binaltorphimine and the peripherally active opioid antagonist naloxone-methiodide were ineffective in blocking the antinociceptive effects of MMP-2200. At equi-antinociceptive doses, MMP-2200 produced significantly less stimulation of locomotor activity compared with morphine. Repeated administration of equivalent doses of morphine and MMP-2200 (twice daily for 3 days) produced antinociceptive tolerance (~13- and 5-fold rightward shifts, respectively). In acute and chronic physical dependence assays, naloxone precipitated a more severe withdrawal in mice receiving morphine compared with equivalent doses of the glycopeptide. Both morphine and MMP-2200 inhibited respiration and gastrointestinal transit. In summary, MMP-2200 acts as a mixed DOR/MOR agonist in vivo, which may in part account for its high antinociceptive potency after systemic administration, as well as its decreased propensity to produce locomotor stimulation, tolerance, and physical dependence in mice, compared with the MOR-selective agonist morphine. For other measures (e.g., gastrointestinal transit and respiration), the significant MOR component may not allow differentiation from morphine.


Assuntos
Analgésicos Opioides/farmacologia , Glicopeptídeos/farmacologia , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Analgésicos Opioides/química , Animais , Células CHO , Cricetinae , Cricetulus , Glicopeptídeos/química , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Atividade Motora/efeitos dos fármacos , Atividade Motora/fisiologia , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Receptores Opioides delta/fisiologia , Receptores Opioides mu/fisiologia
11.
Bioorg Med Chem ; 19(9): 2808-16, 2011 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-21482470

RESUMO

A novel series of homo- and heterodimeric ligands containing κ/µ agonist and µ agonist/antagonist pharmacophores joined by a 10-carbon ester linker chain were synthesized and evaluated for their in vitro binding affinity at κ, µ, and δ opioid receptors, and their functional activities were determined at κ and µ receptors in [(35)S]GTPγS functional assays. Most of these compounds had high binding affinity at µ and κ receptors (K(i) values less than 1nM). Compound 15b, which contains butorphan (1) at one end of linking chain and butorphanol (5) at the other end, was the most potent ligand in this series with binding affinity K(i) values of 0.089nM at the µ receptor and 0.073nM at the κ receptor. All of the morphinan-derived ligands were found to be partial κ and µ agonists; ATPM-derived ligands 12 and 11 were found to be full κ agonists and partial µ agonists.


Assuntos
Decanoatos/síntese química , Morfinanos/química , Receptores Opioides delta/química , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Animais , Butorfanol/química , Células CHO , Cricetinae , Cricetulus , Decanoatos/química , Decanoatos/farmacologia , Humanos , Ligantes , Morfinanos/síntese química , Morfinanos/farmacologia , Ligação Proteica , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/antagonistas & inibidores , Receptores Opioides mu/metabolismo
12.
J Neuroimmune Pharmacol ; 16(4): 699-705, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34935110

RESUMO

This guest commentary introduces "The Neuroimmune Pharmacology of SARS-CoV-2," a special theme issue for The Journal of Neuroimmune Pharmacology led by the Society on NeuroImmune Pharmacology. The issue builds on the Society's Virtual Workshop on COVID-19 held April 9, 2021. Top row from left: Drs. Santosh Kumar, Sowmya Yelamanchili, Pankaj Seth, Jean M. Bidlack; Bottom row from left: Drs. Gurudutt Pendyala, Sanjay Maggirwar, and Sulie L. Chang.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos
13.
J Neuroimmune Pharmacol ; 16(3): 519-530, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34181181

RESUMO

This brief report collects the program and abstracts of the Society on NeuroImmune Pharmacology (SNIP) COVID-19 Virtual Workshop held on April 9, 2021. The workshop consisted of four symposia: Symposium 1: Molecular approaches to COVID-19 pathogenesis and underlying mechanisms; Symposium 2: Therapeutic and vaccine approaches to COVID-19; Symposium 3: Early Career Investigator talks; and Symposium 4: Diversity and Inclusion SNIP Committee (DISC) program: Well-being and reflections. The workshop also featured four special talks on COVID-19 and funding opportunities from the National Institute on Alcohol Abuse and Alcoholism (NIAAA); COVID-19 and funding opportunities from the National Institute on Drug Abuse (NIDA); opportunities from NIH for early career investigator (ECI) fellows; and neurologic and psychiatric complications of SARS-CoV-2 infection. Presenters included NIH officials, SNIP members, and non-member scientists whose abstracts were submitted and accepted for inclusion in the virtual event hosted by the University of Nebraska Medical Center via Zoom webinar. A special theme issue of SNIP's official journal, the Journal of Neuroimmune Pharmacology (JNIP), will collect select papers from the workshop along with other related manuscripts in a special theme issue titled "Neuroimmune Pharmacology of SARS-CoV-2."


Assuntos
Tratamento Farmacológico da COVID-19 , COVID-19/imunologia , Educação/tendências , Neuroimunomodulação/imunologia , Sociedades Científicas/tendências , Antivirais/administração & dosagem , Antivirais/imunologia , Educação/métodos , Humanos , Fatores Imunológicos/administração & dosagem , Fatores Imunológicos/imunologia , Neuroimunomodulação/efeitos dos fármacos
14.
J Neuroimmune Pharmacol ; 16(4): 706-717, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34826061

RESUMO

In spring of 2021, the Society on NeuroImmune Pharmacology (SNIP) organized a virtual workshop on the coronavirus disease 2019 (COVID-19). The daylong event's fourth and final symposium, "Well-being and reflections," offered a glimpse at the pandemic's impact on the lives of our scientists and educators. This manuscript includes a brief summary of the symposium, a transcription of our incoming president Dr. Santosh Kumar's lecture, titled "Intervention and improved well-being of basic science researchers during the COVID-19 era: a case study," and the panel discussion that followed, "Reflection and sharing," featuring Drs. Jean M. Bidlack, Sylvia Fitting, Santhi Gorantla, Maria Cecilia G. Marcondes, Loyda M. Melendez, and Ilker K. Sariyer. The conclusion of this manuscript includes comments from SNIP's president Dr. Sulie L. Chang and our Chief Editor, Dr. Howard E. Gendelman. Drs. Sowmya Yelamanchili and Jeymohan Joseph co-chaired the symposium.


Assuntos
COVID-19 , Humanos , Pandemias , SARS-CoV-2
15.
Bioorg Med Chem Lett ; 20(5): 1507-9, 2010 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-20144870

RESUMO

A series of bivalent hydroxy ether butorphan ligands were prepared and their binding affinities at the opioid receptors determined. Addition of a hydroxy group to a hydrocarbon chain can potentiate binding affinity up to 27- and 86-fold at the mu and kappa opioid receptors, respectively. Two bivalent ligands with sub-nanomolar binding affinity at the mu and kappa opioid receptors were discovered.


Assuntos
Analgésicos/química , Ligantes , Morfinanos/química , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Analgésicos/síntese química , Analgésicos/farmacologia , Animais , Células CHO , Cricetinae , Cricetulus , Morfinanos/síntese química , Morfinanos/farmacologia , Ligação Proteica , Receptores Opioides kappa/antagonistas & inibidores , Receptores Opioides mu/antagonistas & inibidores
17.
Psychopharmacology (Berl) ; 237(4): 1195-1208, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31912192

RESUMO

RATIONALE AND OBJECTIVES: The present study characterized the behavioral pharmacology of a novel, mixed-action delta-selective (78:1) opioid receptor agonist, BBI-11008. This glycopeptide drug candidate was tested in assays assessing antinociception (acute, inflammatory, and neuropathic pain-like conditions) and side-effect endpoints (respiratory depression and drug self-administration). RESULTS: BBI-11008 had a 78-fold greater affinity for the delta opioid receptor than the mu receptor, and there was no binding to the kappa opioid receptor. BBI-11008 (3.2-100; 10-32 mg kg-1, i.v.) and morphine (1-10; 1-3.2 mg kg-1, i.v.) produced antinociceptive and anti-allodynic effects in assays of acute thermal nociception and complete Freund's adjuvant (CFA)-induced inflammatory pain, with BBI-11008 being less potent than morphine in both assays. BBI-11008 (1-18 mg kg-1, i.v.) had similar efficacy to gabapentin (10-56 mg kg-1, i.v.) in a spinal nerve ligation (SNL) model of neuropathic pain. In the respiration assay, with increasing %CO2 exposure, BBI-11008 produced an initial increase (32 mg kg-1, s.c.) and then decrease (56 mg kg-1, s.c.) in minute volume (MV) whereas morphine (3.2-32 mg kg-1, s.c.) produced dose-dependent decreases in MV. In the drug self-administration procedure, BBI-11008 did not maintain self-administration at any dose tested. CONCLUSIONS: These results suggest that the glycopeptide drug candidate possesses broad-spectrum antinociceptive and anti-allodynic activity across a range of pain-like conditions. Relative to morphine or fentanyl, the profile for BBI-11008 in the respiration and drug self-administration assays suggests that BBI-11008 may have less pronounced deleterious side effects. Continued assessment of this compound is warranted.


Assuntos
Analgésicos Opioides/administração & dosagem , Neuralgia/tratamento farmacológico , Medição da Dor/efeitos dos fármacos , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Mecânica Respiratória/efeitos dos fármacos , Analgésicos Opioides/química , Animais , Células CHO , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Humanos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/psicologia , Masculino , Camundongos , Morfina/administração & dosagem , Neuralgia/metabolismo , Neuralgia/psicologia , Medição da Dor/psicologia , Ratos , Ratos Sprague-Dawley , Receptores Opioides delta/metabolismo , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Mecânica Respiratória/fisiologia , Autoadministração
18.
J Neurosci ; 28(47): 12183-9, 2008 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-19020012

RESUMO

The Gbetagamma subunit has been implicated in many downstream signaling events associated with opioids. We previously demonstrated that a small molecule inhibitor of Gbetagamma-subunit-dependent phospholipase (PLC) activation potentiated morphine-induced analgesia (Bonacci et al., 2006). Here, we demonstrate that this inhibitor, M119 (cyclohexanecarboxylic acid [2-(4,5,6-trihydroxy-3-oxo-3H-xanthen-9-yl)-(9Cl)]), is selective for mu-opioid receptor-dependent analgesia and has additional efficacy in mouse models of acute tolerance and dependence. When administered by an intracerebroventricular injection in mice, M119 caused 10-fold and sevenfold increases in the potencies of morphine and the mu-selective peptide, DAMGO, respectively. M119 had little or no effect on analgesia induced by the kappa agonist U50,488 or delta agonists DPDPE or Deltorphin II. Similar results were obtained in vitro, as only activation of the mu-opioid receptor stimulated PLC activation, whereas no effect was seen with the kappa- and delta-opioid receptors. M119 inhibited mu-receptor-dependent PLC activation. In studies to further explore the in vivo efficacy of M119, systemic administration M119 also resulted in a fourfold shift increase in potency of systemically administered morphine. Of particular interest, M119 was also able to attenuate acute, antinociceptive tolerance and dependence in mice treated concomitantly with both M119 and morphine. These studies suggest that small organic molecules, such as M119, that specifically regulate Gbetagamma subunit signaling may have important therapeutic applications in enhancing opioid analgesia, while attenuating the development of tolerance and dependence.


Assuntos
Analgésicos/efeitos adversos , Cicloexanos/uso terapêutico , Tolerância a Medicamentos/fisiologia , Dependência de Morfina/tratamento farmacológico , Morfina/efeitos adversos , Receptores Opioides mu/fisiologia , Xantenos/uso terapêutico , Analgésicos/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Células CHO , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Cicloexanos/química , Cicloexanos/farmacologia , Relação Dose-Resposta a Droga , Interações Medicamentosas , Humanos , Fosfatos de Inositol/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Dependência de Morfina/etiologia , Dependência de Morfina/fisiopatologia , Naloxona/farmacologia , Medição da Dor/métodos , Fosfolipase C beta/farmacologia , Ligação Proteica/efeitos dos fármacos , Tempo de Reação/efeitos dos fármacos , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transfecção/métodos , Xantenos/química , Xantenos/farmacologia
19.
Bioorg Med Chem Lett ; 19(2): 365-8, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19091564

RESUMO

A series of 7,8- and 8,9-fused triazole and imidazole analogues of cyclazocine have been made and characterized in opioid receptor binding and [(35)S]GTPgammaS assays. Target compounds were designed to explore the SAR surrounding our lead molecule for this study, namely the 8,9-fused pyrrolo analogue 2 of cyclazocine. Compared to 2, many of the new compounds in this study displayed very high affinity for opioid receptors.


Assuntos
Ciclazocina/síntese química , Ciclazocina/farmacologia , Receptores Opioides/efeitos dos fármacos , Animais , Células CHO , Cricetinae , Cricetulus , Ciclazocina/análogos & derivados , Ciclazocina/metabolismo , Ciclização , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Ensaio Radioligante , Receptores Opioides/metabolismo , Relação Estrutura-Atividade , Radioisótopos de Enxofre
20.
Bioorg Med Chem Lett ; 19(1): 203-8, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19027293

RESUMO

A series of 15 novel opioid derivatives were made where the prototypic phenolic-OH group of traditional opioids was replaced by a carboxamido (CONH(2)) group. For 2,6-methano-3-benzazocines and morphinans similar or, in a few instances, enhanced affinity for mu, delta and kappa opioid receptors was observed when the OH-->CONH(2) switch was applied. For 4,5alpha-epoxymorphinans, binding affinities for the corresponding carboxamide derivatives were much lower than the OH partner consistent with our pharmacophore hypothesis concerning carboxamide bioactive conformation. The active metabolite of tramadol and its carboxamide counterpart had comparable affinities for the three receptors.


Assuntos
Analgésicos Opioides/síntese química , Receptores Opioides/metabolismo , Amidas , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacologia , Humanos , Ligação Proteica , Relação Estrutura-Atividade , Tramadol/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA