Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Life Sci ; 306: 120788, 2022 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-35817166

RESUMEN

AIMS: We determined the ability of the multi-chemokine receptor (CCR2/CCR5/CCR8) antagonist RAP-103 to modulate pain behaviors in an acute model of surgical pain, with and without an added opioid (morphine), and by itself in a chronic model of Streptozotocin (STZ)-induced diabetic peripheral neuropathy (DPN). MATERIALS AND METHODS: Pain behaviors were assessed by mechanical and thermal tests in rats. Cytokine and chemokine biomarkers in sciatic nerve and spinal cord were assessed by in situ qPCR. KEY FINDINGS: In the incisional pain assay, RAP-103 (0.01-1 mg/kg, i.p.) alone had no antiallodynic effect post-surgery. RAP-103 (0.5 mg/kg) when co-administered with morphine (0.5-5 mg/kg), reduced the ED50 of morphine from 3.19 mg/kg to 1.42 mg/kg. In a DPN model, rats exhibited persistent mechanical and cold allodynia. Oral administration of RAP-103 (0.5-0.02 mg/kg/day) resulted in a complete reversal of established hypersensitivity in DPN rats (P < .001), which gradually returned to pain hypersensitivity after the cessation of the treatment. The mRNA expression of cytokines, IL-1ß, TNFα; chemokines CCL2, CCL3; and chemokine receptors CCR2 and CCR5 in DPN rat sciatic nerve, but not spinal cord, were significantly increased. RAP-103 resulted in significant reductions in sciatic nerve expression of IL-1ß, TNFα and CCL3 in STZ-induced diabetic rats with trends toward lower levels for CCL2 and CCR5, while CCR2 was unchanged. SIGNIFICANCE: In acute pain, co-administration of RAP-103 with morphine provided the same antinociceptive effect with a reduced dose of morphine, reducing opioid side-effects and risks. RAP-103 by itself is an effective non-opioid antinociceptive treatment for diabetic neuropathic pain.


Asunto(s)
Diabetes Mellitus Experimental , Neuropatías Diabéticas , Neuralgia , Animales , Ratas , Analgésicos Opioides/farmacología , Analgésicos Opioides/uso terapéutico , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/tratamiento farmacológico , Neuropatías Diabéticas/tratamiento farmacológico , Hiperalgesia/metabolismo , Morfina/farmacología , Morfina/uso terapéutico , Neuralgia/metabolismo , Péptidos/uso terapéutico , Receptores de Quimiocina , Factor de Necrosis Tumoral alfa
2.
Life Sci ; 285: 120014, 2021 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-34619167

RESUMEN

AIMS: We have shown that chemokines injected into the periaqueductal gray region of the brain blocks opioid-induced analgesia in the rat cold-water tail flick test (CWTF). The present experiments tested whether chemokine receptor antagonists (CRAs), in combination with sub-analgesic doses of morphine, would provide maximal analgesia in the CWTF test and the mouse formalin pain assay. The effect of CRAs on respiratory depression was also evaluated. MAIN METHODS: One, two or four CRAs (AMD3100/CXCR4, maraviroc/CCR5, RS504393/CCR2 orAZD8797/CX3CR1) were used in combination with sub-analgesic doses of morphine, all given systemically. Pain was assessed using the rat CWTF test or formalin injection into the paw of mice scored by licking. Respiration and oxygen saturation were measured in rats using a MouseOX® Plus - pulse oximeter. KEY FINDINGS: In the CWTF test, a sub-maximal dose of morphine in combination with maraviroc alone, maraviroc plus AMD3100, or with the four chemokine receptor antagonists, produced synergistic increases in antinociception. In the formalin test, the combination of four CRAs plus a sub-maximal dose of morphine resulted in increased antinociception in both male and female mice. AMD3100 had an additive effect with morphine in both sexes. Coadministration of CRAs with morphine did not potentiate the opioid respiratory depressive effect. SIGNIFICANCE: These results support the conclusion that combinations of CRAs can increase the potency of sub-analgesic doses of morphine analgesia without increasing respiratory depression. The results support an "opioid sparing" strategy for alleviation of pain using reduced doses of opioids in combination with CRAs to achieve maximal analgesia.


Asunto(s)
Analgesia/métodos , Analgésicos Opioides/farmacología , Morfina/farmacología , Nocicepción/efectos de los fármacos , Dolor Nociceptivo/tratamiento farmacológico , Receptores de Quimiocina/antagonistas & inhibidores , Animales , Bencilaminas/administración & dosificación , Bencilaminas/farmacología , Ciclamas/administración & dosificación , Ciclamas/farmacología , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Femenino , Masculino , Maraviroc/administración & dosificación , Maraviroc/farmacología , Morfina/administración & dosificación , Morfina/efectos adversos , Dolor Nociceptivo/fisiopatología , Pirimidinas/administración & dosificación , Pirimidinas/farmacología , Ratas , Ratas Sprague-Dawley , Insuficiencia Respiratoria/inducido químicamente , Tiazoles/administración & dosificación , Tiazoles/farmacología
3.
Front Pharmacol ; 12: 804950, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35185546

RESUMEN

Previous work from our laboratory showed that a CB2 selective agonist, O-1966, blocked the proliferative response of C57BL/6 mouse spleen cells exposed to spleen cells of C3HeB/FeJ mice in vitro in the mixed lymphocyte reaction (MLR). The MLR is widely accepted as an in vitro correlate of in vivo grant rejection. Mechanisms of the immunosuppression induced by the cannabinoid were explored, and it was shown that O-1966 in this in vitro assay induced CD25+Foxp3+ Treg cells and IL-10, as well as down-regulated mRNA for CD40 and the nuclear form of the transcription factors NF-κB and NFAT in T-cells. The current studies tested the efficacy of O-1966 in prolonging skin grafts in vivo. Full thickness flank skin patches (1-cm2) from C3HeB/FeJ mice were grafted by suturing onto the back of C57BL/6 mice. O-1966 or vehicle was injected intraperitoneally into treated or control groups of animals beginning 1 h pre-op, and then every other day until 14 days post-op. Graft survival was scored based on necrosis and rejection. Treatment with 5 mg/kg of O-1966 prolonged mean graft survival time from 9 to 11 days. Spleens harvested from O-1966 treated mice were significantly smaller than those of vehicle control animals based on weight. Flow cytometry analysis of CD4+ spleen cells showed that O-1966 treated animals had almost a 3-fold increase in CD25+Foxp3+ Treg cells compared to controls. When dissociated spleen cells were placed in culture ex vivo and stimulated with C3HeB/FeJ cells in an MLR, the cells from the O-1966 treated mice were significantly suppressed in their proliferative response to the allogeneic cells. These results support CB2 selective agonists as a new class of compounds to prolong graft survival in transplant patients.

4.
Molecules ; 25(11)2020 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-32517185

RESUMEN

(-)-N-Phenethyl analogs of optically pure N-norhydromorphone were synthesized and pharmacologically evaluated in several in vitro assays (opioid receptor binding, stimulation of [35S]GTPγS binding, forskolin-induced cAMP accumulation assay, and MOR-mediated ß-arrestin recruitment assays). "Body" and "tail" interactions with opioid receptors (a subset of Portoghese's message-address theory) were used for molecular modeling and simulations, where the "address" can be considered the "body" of the hydromorphone molecule and the "message" delivered by the substituent (tail) on the aromatic ring of the N-phenethyl moiety. One compound, N-p-chloro-phenethynorhydromorphone ((7aR,12bS)-3-(4-chlorophenethyl)-9-hydroxy-2,3,4,4a,5,6-hexahydro-1H-4,12-methanobenzofuro[3,2-e]isoquinolin-7(7aH)-one, 2i), was found to have nanomolar binding affinity at MOR and DOR. It was a potent partial agonist at MOR and a full potent agonist at DOR with a δ/µ potency ratio of 1.2 in the ([35S]GTPγS) assay. Bifunctional opioids that interact with MOR and DOR, the latter as agonists or antagonists, have been reported to have fewer side-effects than MOR agonists. The p-chlorophenethyl compound 2i was evaluated for its effect on respiration in both mice and squirrel monkeys. Compound 2i did not depress respiration (using normal air) in mice or squirrel monkeys. However, under conditions of hypercapnia (using air mixed with 5% CO2), respiration was depressed in squirrel monkeys.


Asunto(s)
Hidromorfona/análogos & derivados , Hipercapnia/tratamiento farmacológico , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Animales , Unión Competitiva , Hidromorfona/química , Hidromorfona/farmacología , Hipercapnia/patología , Ratones , Modelos Moleculares , Unión Proteica , Receptores Opioides delta/antagonistas & inhibidores , Receptores Opioides delta/metabolismo , Receptores Opioides mu/antagonistas & inhibidores , Receptores Opioides mu/metabolismo , Respiración Artificial , Saimiri , Relación Estructura-Actividad
5.
Mil Med ; 185(Suppl 1): 130-135, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-32074321

RESUMEN

INTRODUCTION: Although opioids are widely prescribed for pain, in many circumstances, they have only modest efficacy. Preclinical studies have shown that chemokines, immune mediators released during tissue injury and inflammation, can desensitize opioid receptors and block opioid analgesia by a process termed "heterologous desensitization." The present studies tested the hypothesis that in evoked pain, certain chemokine receptor antagonists (CRAs), given with a submaximal dose of morphine, would result in enhanced morphine potency. METHODS: Three rodent pain assays were used: incisional pain in rats, the cold-water tail flick test in rats, and the formalin test in mice. The FDA-approved, commercially available CRAs, maraviroc and AMD3100, were used. They block the chemokine receptors and ligands, CCR5/CCL5 (RANTES) and CXCR4/CXCL4 (SDF-1α), respectively. RESULTS: In the incisional pain assay, it was found that the combination of a single CRA, or of both CRAs, with morphine significantly shifted the morphine dose-response curve to the left, as much as 3.3-fold. In the cold-water tail flick and formalin tests, significant increases of the antinociceptive effects of morphine were also observed when combined with CRAs. CONCLUSIONS: These results support the potential of a new "opioid-sparing" approach for pain treatment, which combines CRAs with reduced doses of morphine.


Asunto(s)
Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Morfina/uso terapéutico , Receptores de Quimiocina/antagonistas & inhibidores , Analgésicos Opioides/farmacología , Analgésicos Opioides/uso terapéutico , Análisis de Varianza , Animales , Bencilaminas , Ciclamas , Modelos Animales de Enfermedad , Compuestos Heterocíclicos/farmacología , Compuestos Heterocíclicos/uso terapéutico , Maraviroc/farmacología , Maraviroc/uso terapéutico , Morfina/farmacología , Manejo del Dolor/métodos , Manejo del Dolor/normas , Manejo del Dolor/estadística & datos numéricos , Dimensión del Dolor/métodos , Ratas , Ratas Sprague-Dawley , Herida Quirúrgica/complicaciones , Herida Quirúrgica/tratamiento farmacológico
6.
Br J Pharmacol ; 176(17): 3378-3389, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31218677

RESUMEN

BACKGROUND AND PURPOSE: Much of the opioid epidemic arose from abuse of prescription opioid drugs. This study sought to determine if the combination of a cannabinoid with an opioid could produce additive or synergistic effects on pain, allowing reduction in the opioid dose needed for maximal analgesia. EXPERIMENTAL APPROACH: Pain was assayed using the formalin test in mice and the carrageenan assay in rats. Morphine and two synthetic cannabinoids were tested: WIN55,212-2 (WIN), which binds to both CB1 and CB2 receptors, and possibly TRPV1 channels; and GP1a, which has activity at CB2 receptors and is reported to inhibit fatty acid amide hydrolase, thus raising levels of endogenous cannabinoids. KEY RESULTS: Morphine in combination with WIN in the formalin test gave synergistic analgesia. Studies with selective antagonists showed that WIN was acting through CB1 receptors. Morphine in combination with GP1a in the formalin test was sub-additive. In the carrageenan test, WIN had no added effect when combined with morphine, but GP1a with morphine showed enhanced analgesia. Both WIN and Gp1a used alone had analgesic activity in the formalin pain test, but not in the carrageenan pain test. CONCLUSIONS AND IMPLICATIONS: The ability of a cannabinoid to produce an additive or synergistic effect on analgesia when combined with morphine varies with the pain assay and may be mediated by CB1 or CB2 receptors. These results hold the promise of using cannabinoids to reduce the dose of opioids for analgesia in certain pain conditions.


Asunto(s)
Analgésicos Opioides/farmacología , Cannabinoides/farmacología , Morfina/farmacología , Dolor/tratamiento farmacológico , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB2/metabolismo , Animales , Carragenina , Relación Dosis-Respuesta a Droga , Formaldehído , Masculino , Ratones , Dolor/inducido químicamente , Dolor/metabolismo , Manejo del Dolor , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad
8.
J Pharmacol Exp Ther ; 367(3): 433-441, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30249618

RESUMEN

Crossdesensitization between opioid and chemokine receptors and involvement of chemokines in pain modulation are well established. We investigated if coadministration of chemokine receptor antagonists (CRAs) with morphine would enhance the analgesic potency of morphine on incisional pain in rats. Animals underwent incisional surgery on the left hind paw and pain responses were evaluated using von Frey filaments at various time points postsurgery between 15 and 360 minutes and daily between 24 and 72 hours. Dose-response curves for morphine, maraviroc (a CCR5 antagonist), and AMD3100 (a CXCR4 antagonist) alone were established. While morphine significantly reduced pain in a time- and dose-dependent manner, maraviroc and AMD3100 had no effect by themselves. Coadministration of either maraviroc or AMD3100 with morphine significantly increased morphine's analgesic effect on incisional pain, shifting the dose-response curve to the left 2.3- and 1.8-fold, respectively. Coadministration of both CRAs with morphine significantly shifted further the morphine dose-response curve to the left 3.3-fold. The effect of treatments on mRNA levels in the draining popliteal lymph node for a panel of chemokines and cytokines showed that message for many of these mediators was upregulated by the incision, and the combination of morphine with the CRAs markedly downregulated them. The data show that combining morphine with CRAs potentiates morphine's analgesic effect on incisional pain. Thus, the same analgesic effect of morphine alone can be achieved with lower doses of morphine when combined with CRAs. Using morphine in lower doses could reduce unwanted side effects and possibly block development of tolerance and dependence.


Asunto(s)
Analgésicos Opioides/farmacología , Morfina/farmacología , Dolor/tratamiento farmacológico , Receptores de Quimiocina/antagonistas & inhibidores , Animales , Regulación hacia Abajo/efectos de los fármacos , Tolerancia a Medicamentos/fisiología , Masculino , Dolor/metabolismo , Dimensión del Dolor/métodos , Ratas , Ratas Sprague-Dawley , Receptores CXCR4/metabolismo
9.
Front Microbiol ; 6: 1230, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26583016

RESUMEN

Although opioids have been extensively studied for their impact on the immune system, limited information is available about the specific actions of opioids on intracellular antiviral innate immunity against HIV infection. Thus, we investigated whether heroin, one of the most abused drugs, inhibits the expression of intracellular HIV restriction microRNA (miRNA) and facilitates HIV replication in macrophages. Heroin treatment of macrophages enhanced HIV replication, which was associated with the downregulation of several HIV restriction miRNAs. These heroin-mediated actions on the miRNAs and HIV could be antagonized by naltrexone, an opioid receptor antagonist. Furthermore, the in vitro negative impact of heroin on HIV-associated miRNAs was confirmed by the in vivo observation that heroin addicts had significantly lower levels of macrophage-derived HIV restriction miRNAs than those in the control subjects. These in vitro and in vivo findings indicate that heroin use compromises intracellular anti-HIV innate immunity, providing a favorable microenvironment for HIV survival in the target cells.

10.
J Neuroimmune Pharmacol ; 10(2): 318-32, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25980325

RESUMEN

We have previously shown that agonists selective for the cannabinoid receptor 2 (CB2), including O-1966, inhibit the Mixed Lymphocyte Reaction (MLR), an in vitro correlate of organ graft rejection, predominantly through effects on T-cells. Current studies explored the mechanism of this immunosuppression by O-1966 using mouse spleen cells. Treatment with O-1966 dose-relatedly decreased levels of the active nuclear forms of the transcription factors NF-κB and NFAT in wild-type T-cells, but not T-cells from CB2 knockout (CB2R k/o) mice. Additionally, a gene expression profile of purified T-cells from MLR cultures generated using a PCR T-cell activation array showed that O-1966 decreased mRNA expression of CD40 ligand and CyclinD3, and increased mRNA expression of Src-like-adaptor 2 (SLA2), Suppressor of Cytokine Signaling 5 (SOCS5), and IL-10. The increase in IL-10 was confirmed by measuring IL-10 protein levels in MLR culture supernatants. Further, an increase in the percentage of regulatory T-cells (Tregs) was observed in MLR cultures. Pretreatment with anti-IL-10 resulted in a partial reversal of the inhibition of proliferation and blocked the increase of Tregs. Additionally, O-1966 treatment caused a dose-related decrease in the expression of CD4 in MLR cultures from wild-type, but not CB2R k/o, mice. These data support the potential of CB2-selective agonists as useful therapeutic agents to prolong graft survival in transplant patients, and strengthens their potential as a new class of immunosuppressive agents with broader applicability.


Asunto(s)
Anisoles/farmacología , Agonistas de Receptores de Cannabinoides/farmacología , Interleucina-10/biosíntesis , Receptor Cannabinoide CB2/agonistas , Receptor Cannabinoide CB2/metabolismo , Linfocitos T Reguladores/metabolismo , Animales , Técnicas de Cocultivo , Ciclohexanoles , Relación Dosis-Respuesta a Droga , Femenino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Linfocitos T Reguladores/efectos de los fármacos
11.
J Neuroimmune Pharmacol ; 8(5): 1239-50, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23824763

RESUMEN

Cannabinoids are known to have anti-inflammatory and immunomodulatory properties. Cannabinoid receptor 2 (CB2) is expressed mainly on leukocytes and is the receptor implicated in mediating many of the effects of cannabinoids on immune processes. This study tested the capacity of Δ(9)-tetrahydrocannabinol (Δ(9)-THC) and of two CB2-selective agonists to inhibit the murine Mixed Lymphocyte Reaction (MLR), an in vitro correlate of graft rejection following skin and organ transplantation. Both CB2-selective agonists and Δ(9)-THC significantly suppressed the MLR in a dose dependent fashion. The inhibition was via CB2, as suppression could be blocked by pretreatment with a CB2-selective antagonist, but not by a CB1 antagonist, and none of the compounds suppressed the MLR when splenocytes from CB2 deficient mice were used. The CB2 agonists were shown to act directly on T-cells, as exposure of CD3(+) cells to these compounds completely inhibited their action in a reconstituted MLR. Further, the CB2-selective agonists completely inhibited proliferation of purified T-cells activated by anti-CD3 and anti-CD28 antibodies. T-cell function was decreased by the CB2 agonists, as an ELISA of MLR culture supernatants revealed IL-2 release was significantly decreased in the cannabinoid treated cells. Together, these data support the potential of this class of compounds as useful therapies to prolong graft survival in transplant patients.


Asunto(s)
Cannabinoides/farmacología , Rechazo de Injerto/metabolismo , Activación de Linfocitos/efectos de los fármacos , Receptor Cannabinoide CB2/metabolismo , Linfocitos T/efectos de los fármacos , Animales , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Rechazo de Injerto/inmunología , Rechazo de Injerto/prevención & control , Etiquetado Corte-Fin in Situ , Activación de Linfocitos/inmunología , Prueba de Cultivo Mixto de Linfocitos , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , ARN Mensajero/análisis , Linfocitos T/inmunología
12.
Drug Alcohol Depend ; 123(1-3): 277-81, 2012 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-22196236

RESUMEN

BACKGROUND: Buprenorphine is an opioid receptor ligand whose mechanism of action is incompletely understood. METHODS: Using Ca(2+) imaging, we assessed the effects of buprenorphine, ß-endorphin, and morphine on cytosolic Ca(2+) concentration [Ca(2+)](i), in rat striatal neurons. RESULTS: Buprenorphine (0.01-1 µM) increased [Ca(2+)](i) in a dose-dependent manner in a subpopulation of rat striatal neurons. The effect of buprenorphine was largely reduced by naloxone, a non-selective opioid receptor antagonist, but not by µ, κ, δ or NOP-selective antagonists. ß-Endorphin (0.1 µM) increased [Ca(2+)](i) with a lower amplitude and slower time course than buprenorphine. Similar to buprenorphine, the effect of ß-endorphin was markedly decreased by naloxone, but not by opioid-selective antagonists. Morphine (0.1-10 µM), did not affect [Ca(2+)](i) in striatal neurons. CONCLUSIONS: Our results suggest that buprenorphine and ß-endorphin act on a distinct type/subtype of plasmalemmal opioid receptors or activate intracellular opioid-like receptor(s) in rat striatal neurons.


Asunto(s)
Buprenorfina/farmacología , Calcio/metabolismo , Cuerpo Estriado/metabolismo , Citosol/metabolismo , Narcóticos/farmacología , Neuronas/metabolismo , Receptores Opioides/agonistas , Animales , Cuerpo Estriado/citología , Cuerpo Estriado/efectos de los fármacos , Citosol/efectos de los fármacos , Colorantes Fluorescentes , Fura-2 , Ligandos , Morfina/farmacología , Naltrexona/análogos & derivados , Naltrexona/farmacología , Antagonistas de Narcóticos/farmacología , Neuronas/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , betaendorfina/farmacología
13.
J Neuroimmune Pharmacol ; 6(4): 551-65, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21826405

RESUMEN

Acinetobacter baumannii is an important nosocomial pathogen in civilian intensive care units. Recently the incidence has increased in wounded military personnel. Morphine is documented in numerous animal studies to be immunosuppressive and to sensitize to infection. The hypotheses were tested that morphine, administered for analgesia in the battlefield, predisposes to Acinetobacter infection, and that the opioid may have an additive or synergistic effect with trauma. To test these hypotheses, an intraperitoneal infection model was established in mice using several Acinetobacter strains. Morphine administered for 48 h by implantation of a slow-release morphine pellet increased mortality compared to animals receiving a placebo pellet, an effect that was blocked by the mu-opioid receptor antagonist, naltrexone. Acinetobacter burdens in the blood, spleens, livers, and lungs of morphine-treated mice, were significantly higher than those in placebo-treated animals, confirming that mortality was due to potentiated growth of the bacteria. There were also elevated levels of pro-inflammatory cytokines in morphine-treated versus placebo-treated mice. Morphine caused a reduction in the total number of cells in the peritoneal cavity, a decrease in the percentage and total numbers of neutrophils, and a decrease in the total number of macrophages. Morphine treatment also suppressed levels of the neutrophil-inducing molecules, IL-17A and KC/CXCL1. However, IL-17A(-/-) mice given morphine were not sensitized to Acintobacter infection to a greater degree than similarly treated wild-type mice. Trauma alone did not sensitize to Acinetobacter infection, and there was no additive effect between morphine and trauma. These results support the hypothesis that morphine potentiates Acinetobacter infection.


Asunto(s)
Infecciones por Acinetobacter/inmunología , Analgésicos Opioides/efectos adversos , Morfina/efectos adversos , Heridas y Lesiones/inmunología , Acinetobacter baumannii/efectos de los fármacos , Animales , Separación Celular , Citocinas/biosíntesis , Citocinas/inmunología , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Inmunosupresores/efectos adversos , Interleucina-17/genética , Interleucina-17/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfina/inmunología , Sepsis/inmunología , Sepsis/microbiología
14.
Brain Behav Immun ; 25(7): 1434-43, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21569838

RESUMEN

Recently, we have shown that morphine's analgesic activity can be attenuated by chemokines, specifically CCL5 and CXCL12. Because the HIV-1 coat protein, glycoprotein 120 (gp120), binds to the same receptors as do CCL5 and CXCL12, experiments were designed to investigate the effect of gp120 in the brain on antinociception induced by morphine in the cold-water (-3°C) tail-flick (CWT) and hot-plate (+54°C) tests. In addition, mu-opioid-receptor-mediated effects in brain periaqueductal grey (PAG) slices were examined with whole-cell patch-clamp recordings. The results showed that (1) pretreatment with gp120 itself (10, 25, 50, 100 or 133 ng, PAG) had no nociceptive effect in the CWT; (2) pretreatment with gp120 (25 or 100 ng) dose-dependently reduced antinociception induced by subcutaneous (sc) injection of morphine (3 or 6 mg/kg) or PAG injection of morphine (100 ng) in the CWT; (3) a PAG injection of gp120 (133 ng), given 30 min before sc injection of morphine (6 mg/kg), similarly reduced morphine antinociception in the hot-plate test; (4) the inhibitory effect of gp120 on morphine-induced antinociception in the CWT was reversed by AMD3100, an antagonist of CXCR4; (5) pretreatment of slices with gp120 (200 pM) prevented morphine (10 µM)-induced hyperpolarization and reduction of input resistance in PAG neurons. Electrophysiology studies paralleled gp120-induced desensitization of a mu-opioid-receptor-mediated response in PAG neurons at the single-cell level. These studies are the first to demonstrate that the analgesic activity of morphine can be reduced by the presence of gp120 in the PAG and that pretreatment with AMD3100 is able to restore the analgesic effects of morphine.


Asunto(s)
Analgésicos Opioides/farmacología , Proteína gp120 de Envoltorio del VIH/farmacología , Morfina/farmacología , Dimensión del Dolor/efectos de los fármacos , Percepción del Dolor/efectos de los fármacos , Analgesia , Animales , Conducta Animal/efectos de los fármacos , Bencilaminas , Frío , Ciclamas , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Compuestos Heterocíclicos/farmacología , Calor , Masculino , Potenciales de la Membrana/efectos de los fármacos , Sustancia Gris Periacueductal/efectos de los fármacos , Sustancia Gris Periacueductal/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores CXCR4/antagonistas & inhibidores , Receptores Opioides mu/metabolismo
15.
Drug Alcohol Depend ; 118(2-3): 497-9, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21600706

RESUMEN

BACKGROUND: We showed recently that elevated brain levels of the chemokine stromal cell-derived growth factor-1α (SDF-1α/CXCL12, a ligand for the human immunodeficiency virus [HIV] co-receptor CXCR4) diminish the antinociceptive effect of morphine, but failed to influence buprenorphine-induced antinociception. AIMS: Because the HIV-1 coat protein, glycoprotein 120 (gp120) T-tropic strain, binds to the same receptor as SDF-1α/CXCL12, the present experiments were designed to investigate the consequence of administering gp120 to rat brain on buprenorphine-induced antinociception in the 54°C hot plate test. For comparative purposes, the effect of gp120 on an equi-antinociceptive dose of methadone was also examined. METHODS: A sterilized stainless-steel C313G guide cannula was implanted into the periaqueductal grey (PAG), a brain region critical for the processing of pain signals, and a primary site of action of many analgesics. Rats were pretreated with gp120, administered into the PAG. RESULTS: The subsequent antinociception associated with methadone was diminished whereas buprenorphine-induced antinociception was unaffected. Buprenorphine thus appears to be a more effective analgesic than methadone in the presence of gp120 in the brain, a condition that is associated with HIV-related pain and infection.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Buprenorfina/uso terapéutico , Proteína gp120 de Envoltorio del VIH/farmacología , Metadona/uso terapéutico , Dolor/tratamiento farmacológico , Analgesia , Animales , Proteína gp120 de Envoltorio del VIH/metabolismo , Calor , Masculino , Dimensión del Dolor , Umbral del Dolor/efectos de los fármacos , Sustancia Gris Periacueductal/efectos de los fármacos , Sustancia Gris Periacueductal/metabolismo , Ratas , Ratas Sprague-Dawley
16.
Drug Alcohol Depend ; 114(2-3): 246-8, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21112161

RESUMEN

Although morphine is often the best option for treating acute and chronic severe pain, its analgesic activity can be blocked in situations in which there are elevated levels of chemokines. Indeed, recently we have shown that elevated brain levels of the chemokine stromal cell-derived growth factor-1alpha (SDF-1α/CXCL12, the ligand of the HIV co-receptor CXCR4) diminish the antinociceptive effect of morphine. The purpose of the present study was to investigate whether such an effect is restricted to morphine or extends to other opioid medications such as buprenorphine. A sterilized stainless-steel C313G guide cannula was implanted into the periaqueductal grey (PAG), a brain region critical to the processing of pain signals, and a primary site of action of many analgesic compounds. The cold-water (-3°C) tail-flick test (CWT) was used to measure antinociception. Rats were pretreated with SDF-1α/CXCL12 administered into the PAG, and the antinociceptive actions of buprenorphine were measured. Direct infusion of SDF-1α/CXCL12 into the PAG failed to alter the antinociceptive action of buprenorphine. The presence of SDF-1α/CXCL12 in the PAG differentially alters the antinociceptive function of opioid medications. While it was able to diminish the antinociception induced by morphine (Adler et al., 2006), SDF-1α/CXCL12 did not affect the buprenorphine-induced antinociception. Buprenorphine appears to be more effective in the presence of high levels of SDF-1α/CXCL12 in the brain (which frequently occurs during neuroinflammatory conditions).


Asunto(s)
Analgésicos Opioides/farmacología , Química Encefálica/efectos de los fármacos , Buprenorfina/farmacología , Quimiocina CXCL12/metabolismo , Dimensión del Dolor/efectos de los fármacos , Animales , Química Encefálica/genética , Quimiocina CXCL12/biosíntesis , Quimiocina CXCL12/genética , Masculino , Dimensión del Dolor/métodos , Ratas , Ratas Sprague-Dawley , Resultado del Tratamiento
17.
Mol Pharmacol ; 78(4): 560-8, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20826425

RESUMEN

Known agonists of the orphan receptor GPR35 are kynurenic acid, zaprinast, 5-nitro-2-(3-phenylproplyamino) benzoic acid, and lysophosphatidic acids. Their relatively low affinities for GPR35 and prominent off-target effects at other pathways, however, diminish their utility for understanding GPR35 signaling and for identifying potential therapeutic uses of GPR35. In a screen of the Prestwick Library of drugs and drug-like compounds, we have found that pamoic acid is a potent GPR35 agonist. Pamoic acid is considered by the Food and Drug Administration as an inactive compound that enables long-acting formulations of numerous drugs, such as the antihelminthics oxantel pamoate and pyrantel pamoate; the psychoactive compounds hydroxyzine pamoate (Vistaril) and imipramine pamoate (Tofranil-PM); and the peptide hormones triptorelin pamoate (Trelstar) and octreotide pamoate (OncoLar). We have found that pamoic acid induces a G(i/o)-linked, GPR35-mediated increase in the phosphorylation of extracellular signal-regulated kinase 1/2, recruitment of ß-arrestin2 to GPR35, and internalization of GPR35. In mice, it attenuates visceral pain perception, indicating an antinociceptive effect, possibly through GPR35 receptors. We have also identified in collaboration with the Sanford-Burnham Institute Molecular Libraries Probe Production Center new classes of GPR35 antagonist compounds, including the nanomolar potency antagonist methyl-5-[(tert-butylcarbamothioylhydrazinylidene)methyl]-1-(2,4-difluorophenyl)pyrazole-4-carboxylate (CID2745687). Pamoic acid and potent antagonists such as CID2745687 present novel opportunities for expanding the chemical space of GPR35, elucidating GPR35 pharmacology, and stimulating GPR35-associated drug development. Our results indicate that the unexpected biological functions of pamoic acid may yield potential new uses for a common drug constituent.


Asunto(s)
Analgésicos/administración & dosificación , Arrestinas/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Naftoles/administración & dosificación , Receptores Acoplados a Proteínas G/metabolismo , Animales , Arrestinas/agonistas , Línea Celular , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Humanos , Masculino , Ratones , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Receptores Acoplados a Proteínas G/agonistas , Renilla , beta-Arrestinas
18.
Microb Pathog ; 49(6): 330-5, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20688146

RESUMEN

Previous studies from our laboratory demonstrated that mice treated with morphine pellets are sensitized to Salmonella enterica, serovar Typhimurium infection. However, the opioid receptor antagonist, naltrexone, only partially blocked the effect of morphine, raising the possibility that the opioid might have some of its effects through a nonopioid receptor. To further clarify whether sensitization to infection is an opioid receptor-dependent phenomenon, µ-opioid receptor knockout (MORKO) mice were used in the present study. Wild-type (WT) and MORKO mice were treated with morphine and their sensitivity to oral Salmonella infection was assessed by mortality, bacterial burdens in gut associated lymphoid tissue and in blood and peritoneal fluid, and by levels of pro-inflammatory cytokines in plasma. MORKO animals treated with morphine were refractory to a sublethal dose of Salmonella, while similar treatment of WT animals resulted in 100% mortality. WT animals treated with morphine had high bacterial loads in all organs tested, while morphine-treated MORKO animals had no culturable Salmonella in any organs. Pro-inflammatory cytokine levels were elevated in morphine-treated WT but not MORKO mice infected with Salmonella. These results provide definitive evidence that the morphine-mediated enhancement of oral Salmonella infection is dependent on the µ-opioid receptor.


Asunto(s)
Susceptibilidad a Enfermedades/inducido químicamente , Morfina/toxicidad , Receptores Opioides mu/efectos de los fármacos , Salmonelosis Animal/microbiología , Salmonella typhimurium/patogenicidad , Animales , Líquido Ascítico/microbiología , Carga Bacteriana , Sangre/microbiología , Citocinas/sangre , Modelos Animales de Enfermedad , Femenino , Tejido Linfoide/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Opioides mu/deficiencia , Análisis de Supervivencia
19.
J Neuroimmune Pharmacol ; 5(1): 143-54, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20119853

RESUMEN

Nociceptin/orphanin FQ (N/OFQ), added in vitro to murine spleen cells in the picomolar range, suppressed antibody formation to sheep red blood cells in a primary and a secondary plaque-forming cell assay. The activity of the peptide was maximal at 10(-12) M, with an asymmetric U-shaped dose-response curve that extended activity to 10(-14) M. Suppression was not blocked by pretreatment with naloxone. Specificity of the suppressive response was shown using affinity-purified rabbit antibodies against two N/OFQ peptides and with a pharmacological antagonist. Antisera against both peptides were active, in a dose-related manner, in neutralizing N/OFQ-mediated immunosuppression, when the peptide was used at concentrations from 10(-12.3) to 10(-11.6) M. In addition, nociceptin given in vivo by osmotic pump for 48 h suppressed the capacity of spleen cells placed ex vivo to make an anti-sheep red blood cell response. These studies show that nociceptin directly inhibits an adaptive immune response, i.e., antibody formation, both in vitro and in vivo.


Asunto(s)
Inmunidad Adaptativa/efectos de los fármacos , Inmunosupresores/farmacología , Péptidos Opioides/farmacología , Inmunidad Adaptativa/inmunología , Animales , Relación Dosis-Respuesta a Droga , Eritrocitos/efectos de los fármacos , Eritrocitos/inmunología , Femenino , Sueros Inmunes/farmacología , Inmunosupresores/administración & dosificación , Inmunosupresores/antagonistas & inhibidores , Técnicas In Vitro , Infusiones Subcutáneas , Ratones , Ratones Endogámicos C3H , Naloxona/farmacología , Antagonistas de Narcóticos/farmacología , Péptidos Opioides/administración & dosificación , Péptidos Opioides/antagonistas & inhibidores , Bazo/efectos de los fármacos , Bazo/inmunología , Nociceptina
20.
J Pharmacol Exp Ther ; 332(2): 549-53, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19906780

RESUMEN

Wasting syndrome is a common complication of HIV infection and is marked by progressive weight loss and weakness, often associated with fever. The mechanisms involved in the pathogenesis of these syndromes are not well defined, and neither are the brain areas involved. The present study tests a new hypothesis: that the preoptic anterior hypothalamus (POAH), the main brain area for thermoregulation and fever, has a role in the pathogenesis of fever induced by glycoprotein 120 (gp120), the surface envelope protein used by the HIV to gain access into immune cells, and that the CXC chemokine receptors (CXCR4) that serve as a coreceptor for HIV entry mediate the effect. A sterilized stainless steel C313G cannula guide was implanted into the POAH, and a biotelemetry system was used to monitor the body temperature (Tb) changes. The administration of gp120 into the POAH induced fever in a dose-dependent manner. To demonstrate possible links between the gp120 and CXCR4 in generating the fever, we pretreated the rats with 1,1'-[1,4-phenylenebis(methylene)]bis[1,4,8,11-tetraazacyclotetradecane] octohydrobromide dihydrate (AMD 3100), an antagonist of stromal cell-derived growth factor (SDF)-1alpha/CXCL12, acting at its receptor, CXCR4, 30 min before administration of gp120. AMD 3100 significantly reduced the gp120-induced fever. The present studies show that the presence of HIV-1 envelope glycoprotein gp120 in the POAH provokes fever via interaction CXCR4 pathway.


Asunto(s)
Fiebre/etiología , Proteína gp120 de Envoltorio del VIH/farmacología , Área Preóptica/efectos de los fármacos , Receptores CXCR4/antagonistas & inhibidores , Animales , Fármacos Anti-VIH/farmacología , Bencilaminas , Temperatura Corporal/efectos de los fármacos , Ciclamas , Fiebre/inducido químicamente , Proteína gp120 de Envoltorio del VIH/administración & dosificación , Compuestos Heterocíclicos/farmacología , Masculino , Microinyecciones , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...