Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Proc Natl Acad Sci U S A ; 113(44): 12544-12549, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27791151

RESUMO

Chemotherapy-induced peripheral neuropathic pain (CIPNP) is a severe dose- and therapy-limiting side effect of widely used cytostatics that is particularly difficult to treat. Here, we report increased expression of the cytochrome-P450-epoxygenase CYP2J6 and increased concentrations of its linoleic acid metabolite 9,10-EpOME (9,10-epoxy-12Z-octadecenoic acid) in dorsal root ganglia (DRGs) of paclitaxel-treated mice as a model of CIPNP. The lipid sensitizes TRPV1 ion channels in primary sensory neurons and causes increased frequency of spontaneous excitatory postsynaptic currents in spinal cord nociceptive neurons, increased CGRP release from sciatic nerves and DRGs, and a reduction in mechanical and thermal pain hypersensitivity. In a drug repurposing screen targeting CYP2J2, the human ortholog of murine CYP2J6, we identified telmisartan, a widely used angiotensin II receptor antagonist, as a potent inhibitor. In a translational approach, administration of telmisartan reduces EpOME concentrations in DRGs and in plasma and reverses mechanical hypersensitivity in paclitaxel-treated mice. We therefore suggest inhibition of CYP2J isoforms with telmisartan as a treatment option for paclitaxel-induced neuropathic pain.


Assuntos
Benzimidazóis/farmacologia , Benzoatos/farmacologia , Sistema Enzimático do Citocromo P-450/genética , Neuralgia/prevenção & controle , Paclitaxel/farmacologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Animais , Antineoplásicos Fitogênicos/farmacologia , Antineoplásicos Fitogênicos/toxicidade , Citocromo P-450 CYP2J2 , Sistema Enzimático do Citocromo P-450/metabolismo , Feminino , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Ácidos Linoleicos/sangue , Ácidos Linoleicos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Terapia de Alvo Molecular/métodos , Neuralgia/induzido quimicamente , Paclitaxel/toxicidade , Limiar da Dor/efeitos dos fármacos , Telmisartan
2.
Anesth Analg ; 125(5): 1714-1732, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29049116

RESUMO

Opioids represent an efficacious therapeutic modality for some, but not all pain states. Singular reliance on opioid therapy for pain management has limitations, and abuse potential has deleterious consequences for patient and society. Our understanding of pain biology has yielded insights and opportunities for alternatives to conventional opioid agonists. The aim is to have efficacious therapies, with acceptable side effect profiles and minimal abuse potential, which is to say an absence of reinforcing activity in the absence of a pain state. The present work provides a nonexclusive overview of current drug targets and potential future directions of research and development. We discuss channel activators and blockers, including sodium channel blockers, potassium channel activators, and calcium channel blockers; glutamate receptor-targeted agents, including N-methyl-D-aspartate, α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid, and metabotropic receptors. Furthermore, we discuss therapeutics targeted at γ-aminobutyric acid, α2-adrenergic, and opioid receptors. We also considered antagonists of angiotensin 2 and Toll receptors and agonists/antagonists of adenosine, purine receptors, and cannabinoids. Novel targets considered are those focusing on lipid mediators and anti-inflammatory cytokines. Of interest is development of novel targeting strategies, which produce long-term alterations in pain signaling, including viral transfection and toxins. We consider issues in the development of druggable molecules, including preclinical screening. While there are examples of successful translation, mechanistically promising preclinical candidates may unexpectedly fail during clinical trials because the preclinical models may not recapitulate the particular human pain condition being addressed. Molecular target characterization can diminish the disconnect between preclinical and humans' targets, which should assist in developing nonaddictive analgesics.


Assuntos
Analgésicos não Narcóticos/uso terapêutico , Analgésicos Opioides/uso terapêutico , Descoberta de Drogas/métodos , Transtornos Relacionados ao Uso de Opioides/prevenção & controle , Dor/prevenção & controle , Pesquisa Translacional Biomédica/métodos , Analgésicos não Narcóticos/efeitos adversos , Analgésicos não Narcóticos/química , Analgésicos Opioides/efeitos adversos , Analgésicos Opioides/química , Animais , Composição de Medicamentos , Humanos , Terapia de Alvo Molecular , Transtornos Relacionados ao Uso de Opioides/diagnóstico , Transtornos Relacionados ao Uso de Opioides/etiologia , Dor/etiologia , Dor/fisiopatologia , Dor/psicologia , Percepção da Dor/efeitos dos fármacos , Limiar da Dor/efeitos dos fármacos , Fenótipo , Fatores de Risco , Transdução de Sinais/efeitos dos fármacos
4.
Proc Natl Acad Sci U S A ; 108(12): 5098-103, 2011 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-21385944

RESUMO

Numerous G protein-coupled receptors (GPCRs) have been shown to form heteromeric receptors in cell-based assays. Among the many heteromers reported in the opioid receptor family are µ/κ, κ/δ, and µ/δ. However, the in vivo physiological and behavioral relevance for the proposed heteromers have not yet been established. Here we report a unique example of a ligand, N-naphthoyl-ß-naltrexamine (NNTA) that selectively activates heteromeric µ/κ-opioid receptors in HEK-293 cells and induces potent antinociception in mice. NNTA was an exceptionally potent agonist in cells expressing µ/κ-opioid receptors. Intriguingly, it was found to be a potent antagonist in cells expressing only µ-receptors. In the mouse tail-flick assay, intrathecal (i.t.) NNTA produced antinociception that was ~100-fold greater than by intracerebroventricular (i.c.v.) administration. The κ-antagonist, norBNI, decreased the i.t. potency, and the activity was virtually abolished in µ-opioid receptor knockout mice. No tolerance was induced i.t., but marginal tolerance (3-fold) was observed via the i.c.v. route. Moreover, NNTA produced neither significant physical dependence nor place preference in the ED50 dose range. Taken together, this work provides an important pharmacologic tool for investigating the in vivo functional relevance of heteromeric µ/κ-opioid receptors and suggests an approach to potent analgesics with fewer deleterious side effects.


Assuntos
Analgésicos/farmacologia , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Animais , Avaliação Pré-Clínica de Medicamentos , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Camundongos Knockout , Naltrexona/efeitos adversos , Antagonistas de Entorpecentes/efeitos adversos , Antagonistas de Entorpecentes/farmacologia , Receptores Opioides kappa/genética , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo
5.
Commun Biol ; 6(1): 1228, 2023 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-38052846

RESUMO

TRPV1 is an ion channel that transduces noxious heat and chemical stimuli and is expressed in small fiber primary sensory neurons that represent almost half of skin nerve terminals. Tissue injury and inflammation result in the sensitization of TRPV1 and sustained activation of TRPV1 can lead to cellular toxicity though calcium influx. To identify signals that trigger TRPV1 sensitization after a 24-h exposure, we developed a phenotypic assay in mouse primary sensory neurons and performed an unbiased screen with a compound library of 480 diverse bioactive compounds. Chemotherapeutic agents, calcium ion deregulators and protein synthesis inhibitors were long-acting TRPV1 sensitizers. Amongst the strongest TRPV1 sensitizers were proteasome inhibitors, a class that includes bortezomib, a chemotherapeutic agent that causes small fiber neuropathy in 30-50% of patients. Prolonged exposure of bortezomib produced a TRPV1 sensitization that lasted several days and neurite retraction in vitro and histological and behavioral changes in male mice in vivo. TRPV1 knockout mice were protected from epidermal nerve fiber loss and a loss of sensory discrimination after bortezomib treatment. We conclude that long-term TRPV1 sensitization contributes to the development of bortezomib-induced neuropathy and the consequent loss of sensation, major deficits experienced by patients under this chemotherapeutic agent.


Assuntos
Cálcio , Canais de Cátion TRPV , Humanos , Camundongos , Masculino , Animais , Bortezomib/efeitos adversos , Bortezomib/metabolismo , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo , Cálcio/metabolismo , Pele/metabolismo , Camundongos Knockout
6.
Sci Adv ; 7(14)2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33811065

RESUMO

Humans and mice with natural red hair have elevated basal pain thresholds and an increased sensitivity to opioid analgesics. We investigated the mechanisms responsible for higher nociceptive thresholds in red-haired mice resulting from a loss of melanocortin 1 receptor (MC1R) function and found that the increased thresholds are melanocyte dependent but melanin independent. MC1R loss of function decreases melanocytic proopiomelanocortin transcription and systemic melanocyte-stimulating hormone (MSH) levels in the plasma of red-haired (Mc1re/e ) mice. Decreased peripheral α-MSH derepresses the central opioid tone mediated by the opioid receptor OPRM1, resulting in increased nociceptive thresholds. We identified MC4R as the MSH-responsive receptor that opposes OPRM1 signaling and the periaqueductal gray area in the brainstem as a central area of opioid/melanocortin antagonism. This work highlights the physiologic role of melanocytic MC1R and circulating melanocortins in the regulation of nociception and provides a mechanistic framework for altered opioid signaling and pain sensitivity in red-haired individuals.


Assuntos
Analgésicos Opioides , Nociceptividade , Animais , Cabelo , Hormônios Estimuladores de Melanócitos/farmacologia , Camundongos , Receptor Tipo 1 de Melanocortina/genética , Receptor Tipo 4 de Melanocortina/genética
7.
Bioorg Med Chem Lett ; 19(24): 6978-80, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19892550

RESUMO

In an effort to develop antagonists for kappa-mu opioid receptor heterodimers, a series of bivalent ligands 3-6 containing kappa- and mu-antagonist pharmacophores were designed and synthesized. Evaluation of the series in HEK-293 cells revealed 4 (KMN-21) to selectively antagonize the activation of kappa-mu heterodimers, suggesting possible bridging of receptors when the bivalent ligand spacer contains 21 atoms.


Assuntos
Guanidinas/química , Naltrexona/análogos & derivados , Receptores Opioides kappa/antagonistas & inibidores , Receptores Opioides mu/antagonistas & inibidores , Linhagem Celular , Guanidinas/síntese química , Guanidinas/farmacologia , Humanos , Ligantes , Naltrexona/síntese química , Naltrexona/química , Naltrexona/farmacologia
8.
Nat Rev Drug Discov ; 16(11): 810, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28983099

RESUMO

This corrects the article DOI: 10.1038/nrd.2017.87.

9.
Nat Rev Drug Discov ; 16(8): 545-564, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28596533

RESUMO

Acute and chronic pain complaints, although common, are generally poorly served by existing therapies. This unmet clinical need reflects a failure to develop novel classes of analgesics with superior efficacy, diminished adverse effects and a lower abuse liability than those currently available. Reasons for this include the heterogeneity of clinical pain conditions, the complexity and diversity of underlying pathophysiological mechanisms, and the unreliability of some preclinical pain models. However, recent advances in our understanding of the neurobiology of pain are beginning to offer opportunities for developing novel therapeutic strategies and revisiting existing targets, including modulating ion channels, enzymes and G-protein-coupled receptors.


Assuntos
Dor Aguda/tratamento farmacológico , Analgésicos/farmacologia , Dor Crônica/tratamento farmacológico , Dor Aguda/fisiopatologia , Analgésicos/efeitos adversos , Animais , Dor Crônica/fisiopatologia , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Transtornos Relacionados ao Uso de Substâncias/prevenção & controle
10.
Neuron ; 89(5): 956-70, 2016 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-26898779

RESUMO

The regenerative capacity of the injured CNS in adult mammals is severely limited, yet axons in the peripheral nervous system (PNS) regrow, albeit to a limited extent, after injury. We reasoned that coordinate regulation of gene expression in injured neurons involving multiple pathways was central to PNS regenerative capacity. To provide a framework for revealing pathways involved in PNS axon regrowth after injury, we applied a comprehensive systems biology approach, starting with gene expression profiling of dorsal root ganglia (DRGs) combined with multi-level bioinformatic analyses and experimental validation of network predictions. We used this rubric to identify a drug that accelerates DRG neurite outgrowth in vitro and optic nerve outgrowth in vivo by inducing elements of the identified network. The work provides a functional genomics foundation for understanding neural repair and proof of the power of such approaches in tackling complex problems in nervous system biology.


Assuntos
Axônios/fisiologia , Gânglios Espinais/citologia , Regeneração Nervosa/fisiologia , Neurônios/citologia , Doenças do Sistema Nervoso Periférico/fisiopatologia , Animais , Animais Recém-Nascidos , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Moléculas de Adesão Celular/genética , Moléculas de Adesão Celular/metabolismo , Células Cultivadas , Imunoprecipitação da Cromatina , Modelos Animais de Doenças , Regulação da Expressão Gênica/genética , Glutamina-Frutose-6-Fosfato Transaminase (Isomerizante) , Canais Iônicos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos , Regeneração Nervosa/genética , Transferases de Grupos Nitrogenados/genética , Transferases de Grupos Nitrogenados/metabolismo , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo
11.
Life Sci ; 92(8-9): 438-45, 2013 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-22771696

RESUMO

G protein coupled receptors (GPCRs) represent the largest family of cell surface receptors that are involved in regulating several physiological and behavioral responses in organisms. Indeed, over half of all the approved drugs on the market target GPCRs. Over the past twenty years, several lines of evidence have suggested that GPCRs associate to form oligomeric structures that substantially expand the complexity of signaling processes in vivo. In addition, GPCRs have also been shown to functionally regulate ion channels and help fine-tune neurotransmission. In this review, we will discuss recent advances in both mechanisms, with specific focus on opioid receptors, cannabinoid receptors and transient receptor potential (TRP) calcium channels in nociception. A better understanding of such mechanisms will be imperative in designing analgesics devoid of deleterious side effects and mitigating drug abuse.


Assuntos
Nociceptividade/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Canais de Potencial de Receptor Transitório/fisiologia , Analgésicos/farmacologia , Animais , Humanos , Ligantes , Nociceptividade/efeitos dos fármacos , Receptores de Canabinoides/efeitos dos fármacos , Receptores de Canabinoides/fisiologia , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Receptores Opioides/efeitos dos fármacos , Receptores Opioides/fisiologia , Canais de Potencial de Receptor Transitório/efeitos dos fármacos
12.
ACS Chem Biol ; 8(7): 1412-6, 2013 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-23675763

RESUMO

Bivalent ligands that contain two pharmacophores linked by a spacer are promising tools to investigate the pharmacology of opioid receptor heteromers. Evidence for occupation of neighboring protomers by two phamacophores of a single bivalent ligand (bridging) has relied mainly on pharmacological data. In the present study, we have employed an immunocytochemical correlate to support in vivo biological studies that are consistent with bridging. We show that a bivalent mu agonist/delta antagonist (MDAN-21) that is devoid of tolerance due to possible bridging of mu and delta protomers prevents endocytosis of the heteromeric receptors in HEK-293 cells. Conversely, a bivalent ligand (MDAN-16) with a short spacer or monovalent mu agonist give rise to robust internalization. The data suggest that the immobilization of proximal mu and delta protomers is due to bridging by MDAN-21. The finding that MDAN-21 and its shorter spacer homologue MDAN-16 possess equivalent activity in HEK-293 cells, but produce dramatically divergent internalization of mu-delta heteromer, is relevant to the role of internalization and tolerance.


Assuntos
Regiões Promotoras Genéticas/genética , Receptores Opioides delta/química , Receptores Opioides mu/química , Células HEK293 , Humanos , Ácidos Nucleicos Imobilizados , Imuno-Histoquímica , Ligantes , Microscopia Confocal , Modelos Biológicos , Naltrexona/análogos & derivados , Naltrexona/química , Naltrexona/farmacologia , Receptores Opioides delta/genética , Receptores Opioides mu/genética
13.
J Med Chem ; 56(13): 5505-13, 2013 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-23734559

RESUMO

Given that µ opioid (MOP) and canabinoid (CB1) receptors are colocalized in various regions of the central nervous system and have been reported to associate as heteromer (MOP-CB1) in cultured cells, the possibility of functional, endogenous MOP-CB1 in nociception and other pharmacologic effects has been raised. As a first step in investigating this possibility, we have synthesized a series of bivalent ligands 1-5 that contain both µ agonist and CB1 antagonist pharmacophores for use as tools to study the functional interaction between MOP and CB1 receptors in vivo. Immunofluorescent studies on HEK293 cells coexpressing both receptors suggested 5 (20-atom spacer) to be the only member of the series that bridges the protomers of the heteromer. Antinociceptive testing in mice revealed 5 to be the most potent member of the series. As neither a mixture of monovalent ligands 9 + 10 nor bivalents 2-5 produced tolerance in mice, MOR-CB1 apparently is not an important target for reducing tolerance.


Assuntos
Analgésicos Opioides/farmacologia , Receptor CB1 de Canabinoide/antagonistas & inibidores , Receptores Opioides mu/agonistas , Analgésicos Opioides/síntese química , Analgésicos Opioides/química , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Desenho de Fármacos , Tolerância a Medicamentos , Endocitose/efeitos dos fármacos , Imunofluorescência , Células HEK293 , Humanos , Injeções Intraventriculares , Injeções Espinhais , Ligantes , Masculino , Camundongos Endogâmicos ICR , Modelos Químicos , Estrutura Molecular , Dor/fisiopatologia , Dor/prevenção & controle , Receptor CB1 de Canabinoide/genética , Receptor CB1 de Canabinoide/metabolismo , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Relação Estrutura-Atividade
14.
ACS Chem Neurosci ; 3(9): 720-7, 2012 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-23019498

RESUMO

Morphine and related drugs are widely employed as analgesics despite the side effects associated with their use. Although morphine is thought to mediate analgesia through mu opioid receptors, delta opioid receptors have been implicated in mediating some side effects such as tolerance and dependence. Here we present evidence in rhesus monkeys that morphine, fentanyl, and possibly methadone selectively activate mu-delta heteromers to produce antinociception that is potently antagonized by the delta opioid receptor antagonist, naltrindole (NTI). Studies with HEK293 cells expressing mu-delta heteromeric opioid receptors exhibit a similar antagonism profile of receptor activation in the presence of NTI. In mice, morphine was potently inhibited by naltrindole when administered intrathecally, but not intracerebroventricularly, suggesting the possible involvement of mu-delta heteromers in the spinal cord of rodents. Taken together, these results strongly suggest that, in primates, mu-delta heteromers are allosterically coupled and mediate the antinociceptive effects of three clinically employed opioid analgesics that have been traditionally viewed as mu-selective. Given the known involvement of delta receptors in morphine tolerance and dependence, our results implicate mu-delta heteromers in mediating both antinociception and these side effects in primates. These results open the door for further investigation in humans.


Assuntos
Analgésicos Opioides/farmacologia , Analgésicos/farmacologia , Receptores Opioides delta/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos , Analgésicos/química , Analgésicos Opioides/química , Animais , Sinalização do Cálcio/efeitos dos fármacos , Interpretação Estatística de Dados , Feminino , Fentanila/administração & dosagem , Fentanila/farmacologia , Células HEK293 , Humanos , Injeções Espinhais , Macaca mulatta , Masculino , Metadona/administração & dosagem , Metadona/farmacologia , Camundongos , Naltrexona/administração & dosagem , Naltrexona/análogos & derivados , Naltrexona/farmacologia , Antagonistas de Entorpecentes/administração & dosagem , Antagonistas de Entorpecentes/farmacologia , Medição da Dor/efeitos dos fármacos
15.
ACS Chem Neurosci ; 3(7): 505-9, 2012 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-22860219

RESUMO

Coexpressed and colocalized µ- and δ-opioid receptors have been established to exist as heteromers in cultured cells and in vivo. However the biological significance of opioid receptor heteromer activation is less clear. To explore this significance, the efficacy of selective activation of opioid receptors by SNC80 was assessed in vitro in cells singly and coexpressing opioid receptors using a chimeric G-protein-mediated calcium fluorescence assay, SNC80 produced a substantially more robust response in cells expressing µ-δ heteromers than in all other cell lines. Intrathecal SNC80 administration in µ- and δ-opioid receptor knockout mice produced diminished antinociceptive activity compared with wild type. The combined in vivo and in vitro results suggest that SNC80 selectively activates µ-δ heteromers to produce maximal antinociception. These data contrast with the current view that SNC80 selectively activates δ-opioid receptor homomers to produce antinociception. Thus, the data suggest that heteromeric µ-δ receptors should be considered as a target when SNC80 is employed as a pharmacological tool in vivo.


Assuntos
Analgésicos Opioides/farmacologia , Benzamidas/farmacologia , Piperazinas/farmacologia , Receptores Opioides delta/agonistas , Receptores Opioides delta/metabolismo , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Animais , Benzamidas/química , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Medição da Dor/efeitos dos fármacos , Medição da Dor/métodos , Piperazinas/química
16.
ACS Chem Neurosci ; 1(2): 146-54, 2010 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-22816017

RESUMO

Research in the opioid field has relied heavily on the use of standard agonist ligands such as morphine, [d-Ala(2)-MePhe(4)-Glyol(5)]enkephalin (DAMGO), U69593, bremazocine, [d-Pen(2)d-Pen(5)]enkephalin (DPDPE), and deltorphin-II as tools for investigating the three major types of opioid receptors, MOP (µ), KOP (κ), and DOP (δ), that mediate antinociception. The functional selectivity of these ligands has been based on the assumption that opioid receptors exist as homomers. As numerous studies in cultured cells have suggested that opioid receptors can associate both as homomers and heteromers, we have investigated the selectivity of these standard ligands using intracellular calcium release and [(35)S]GTPγS assays in HEK-293 cells that contain singly and coexpressed opioid receptors. The present study reveals that morphine and DAMGO, traditionally classified as µ selective agonists, selectively activate µ-δ heteromeric opioid receptors with greater efficacy than homomeric opioid receptors. Moreover, standard ligands that have been widely employed as κ- and δ-selective agonists display little or no differences in the activation of homomeric and heteromeric opioid receptors. The far-reaching implications of these results are discussed.


Assuntos
Analgésicos Opioides/farmacologia , Ala(2)-MePhe(4)-Gly(5)-Encefalina/farmacologia , Morfina/farmacologia , Receptores Opioides/agonistas , Área Sob a Curva , Benzenoacetamidas/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , D-Penicilina (2,5)-Encefalina/farmacologia , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Células HEK293 , Humanos , Imuno-Histoquímica , Ligantes , Pirrolidinas/farmacologia , Receptores Opioides delta/agonistas , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas
17.
Proc Natl Acad Sci U S A ; 104(14): 6061-5, 2007 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-17389363

RESUMO

Sickle cell anemia is a common genetic disorder in African Americans. Opioid analgesics are traditionally the treatment for the severe pain associated with this disease. Here we reveal that the opioid antagonist naloxone possesses potent analgesic activity in two transgenic mouse models of sickle cell anemia (NY1DD and hBERK1) and not in their respective controls (ICR-CD1 and C57BL/6J) when administered by three parenteral routes [intracerebroventricular (i.c.v.), intrathecal, and subcutaneous]. In the NY1DD mice, naloxone (i.c.v.) possessed approximately 300-fold greater potency than morphine (i.c.v.). Other opioid antagonists (naltrexone, norbinaltorphimine, and naltrindole) were substantially less effective in producing analgesia. Naloxone and morphine were synergistic in NY1DD mice, suggesting different receptor systems. Microarray analysis suggested naloxone-induced down-regulation of the CC chemokine receptor (CCR)5 in NY1DD mice but not in control mice. Pretreatment of control mice with CC chemokine ligand 5 [CCL5 (RANTES)] enabled naloxone to produce analgesia similar to that observed in NY1DD mice. Mu opioid receptor knockout mice treated similarly also displayed analgesia. That the effect of CCL5 was specifically related to CCR5 and/or CCR1 activation was demonstrated by antagonism of analgesia with the chemokine antagonist methionylated RANTES. Similar antagonism of naloxone-induced analgesia also was observed when NY1DD mice were pretreated with methionylated RANTES. These results indicate that CCR5/CCR1 receptors are directly or indirectly involved in analgesia produced by naloxone. The present study suggests that naloxone may be clinically useful in the treatment of pain associated with sickle cell disease and other disorders involving inflammation.


Assuntos
Analgésicos/farmacologia , Anemia Falciforme/tratamento farmacológico , Modelos Animais de Doenças , Naloxona/farmacologia , Dor/tratamento farmacológico , Analgésicos/administração & dosagem , Analgésicos Opioides/farmacologia , Anemia Falciforme/fisiopatologia , Animais , Quimiocina CCL5/farmacologia , Relação Dose-Resposta a Droga , Regulação para Baixo , Sinergismo Farmacológico , Injeções Intraventriculares , Injeções Espinhais , Injeções Subcutâneas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Knockout , Camundongos Transgênicos , Morfina/farmacologia , Naloxona/administração & dosagem , Dor/genética , Dor/metabolismo , Medição da Dor/efeitos dos fármacos , Receptores CCR5/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA