Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 175
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 120(52): e2304900120, 2023 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-38109529

RESUMEN

Diacylglycerol lipase-beta (DAGLß) serves as a principal 2-arachidonoylglycerol (2-AG) biosynthetic enzyme regulating endocannabinoid and eicosanoid metabolism in immune cells including macrophages and dendritic cells. Genetic or pharmacological inactivation of DAGLß ameliorates inflammation and hyper-nociception in preclinical models of pathogenic pain. These beneficial effects have been assigned principally to reductions in downstream proinflammatory lipid signaling, leaving alternative mechanisms of regulation largely underexplored. Here, we apply quantitative chemical- and phospho-proteomics to find that disruption of DAGLß in primary macrophages leads to LKB1-AMPK signaling activation, resulting in reprogramming of the phosphoproteome and bioenergetics. Notably, AMPK inhibition reversed the antinociceptive effects of DAGLß blockade, thereby directly supporting DAGLß-AMPK crosstalk in vivo. Our findings uncover signaling between endocannabinoid biosynthetic enzymes and ancient energy-sensing kinases to mediate cell biological and pain responses.


Asunto(s)
Endocannabinoides , Glicéridos , Humanos , Endocannabinoides/metabolismo , Glicéridos/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Lipoproteína Lipasa/metabolismo , Ácidos Araquidónicos/metabolismo , Dolor
2.
J Pharmacol Exp Ther ; 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38262742

RESUMEN

MJN110 inhibits the enzyme monoacylglycerol lipase (MAGL) to increase levels of the endocannabinoid (eCB) 2-arachidonoylglycerol (2-AG), an endogenous high-efficacy agonist of cannabinoid 1 and 2 receptors (CB1/2R). MAGL inhibitors are under consideration as candidate analgesics, and we reported previously that acute MJN110 produced partial antinociception in an assay of pain-related behavioral depression in mice. Given the need for repeated analgesic administration in many pain patients and the potential for analgesic tolerance during repeated treatment, this study examined antinociceptive effects of repeated MJN110 on pain-related behavioral depression and CB1R-mediated G-protein function. Male and female ICR mice were treated daily for 7 days in a 2x2 design with (a) 1.0 mg/kg/day MJN110or its vehicle followed by (b) intraperitoneal injection of dilute lactic acid (IP acid) or its vehicle as a visceral noxious stimulus to depress nesting behavior. After behavioral testing, G-protein activity was assessed in lumbar spinal cord andfive brain regions using an assay of CP55,940-stimulated [35S]GTPÉ£S activation. As reported previously, acute MJN110 produced partial but significant relief of IP acid-induced nesting depression on Day 1. After 7 days, MJN110 continued to produce significant but partial antinociception in males, while antinociceptive tolerance developed in females. Repeated MJN110 also produced modest decreases in maximum levels of CP55,940-induced [35S]GTPÉ£S binding in spinal cord and most brain regions. These results indicate that repeated treatment with a relatively low antinociceptive MJN110 dose produces only partial and sex-dependent transient antinociception associated with the emergence of CB1R desensitization in this model of IP acid-induced nesting depression. Significance Statement The drug MJN110 inhibits monoacylglycerol lipase (MAGL) to increase levels of the endogenous cannabinoid 2-arachidonoylglycerol and produce potentially useful therapeutic effects including analgesia. This study used an assay of pain-related behavioral depression in mice to show that repeated MJN110 treatment produced (1) weak but sustained antinociception in male mice, (2) antinociceptive tolerance in females, and (3) modest cannabinoid-receptor desensitization that varied by region and sex. Antinociceptive tolerance may limit the utility of MJN110 for treatment of pain.

3.
Mol Pharmacol ; 103(4): 230-240, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36702548

RESUMEN

Cisplatin is a potent first-line therapy for many solid malignancies, such as breast, ovarian, lung, testicular, and head and neck cancer. However, acute kidney injury (AKI) is a major dose-limiting toxicity in cisplatin therapy, which often hampers the continuation of cisplatin treatment. The endocannabinoid system, consisting of anandamide (AEA) and 2-arachidonoylglycerol and cannabinoid receptors, participates in different kidney diseases. Inhibition of fatty acid amide hydrolase (FAAH), the primary enzyme for the degradation of AEA and AEA-related N-acylethanolamines, elicits anti-inflammatory effects; however, little is known about its role in cisplatin nephrotoxicity. The current study tested the hypothesis that genetic deletion of Faah mitigates cisplatin-induced AKI. Male wild-type C57BL6 (WT) and Faah-/- mice were administered a single dose of intraperitoneal injection of cisplatin (30 mg/kg) and euthanatized 72 hours later. Faah-/- mice showed a reduction of cisplatin-induced blood urea nitrogen, plasma creatinine levels, kidney injury markers, and tubular damage in comparison with WT mice. The renal protection from Faah deletion was associated with enhanced tone of AEA-related N-acylethanolamines (palmitoylethanolamide and oleoylethanolamide), attenuated nuclear factor-κB/p65 activity, DNA damage markers p53 and p21, and decreased expression of the inflammatory cytokine interleukin-1ß, as well as infiltration of macrophages and leukocytes in the kidneys. Notably, a selective FAAH inhibitor (PF-04457845) did not interfere with or perturb the antitumor effects of cisplatin in two head and neck squamous cell carcinoma cell lines, HN30 and HN12. Our work highlights that FAAH inactivation prevents cisplatin-induced nephrotoxicity in mice and that targeting FAAH could provide a novel strategy to mitigate cisplatin-induced nephrotoxicity. SIGNIFICANCE STATEMENT: Mice lacking the Faah gene are protected from cisplatin-induced inflammation, DNA damage response, tubular damage, and kidney dysfunction. Inactivation of FAAH could be a potential strategy to mitigate cisplatin-induced nephrotoxicity.


Asunto(s)
Lesión Renal Aguda , Cisplatino , Animales , Masculino , Ratones , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/genética , Endocannabinoides/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados
4.
J Sep Sci ; 46(22): e2300395, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37688356

RESUMEN

Interest has increased in the role of N-acyl amino acids in a variety of disease states and as potential pharmacotherapies. Recently, N-oleoyl glycine and N-oleoyl alanine have shown promise in reducing the rewarding effects of drugs of abuse and alleviating withdrawal signs in rodent models. Previously published methods for the quantitation of these analytes by high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) in tissue were part of extensive lipidomic panels which may result in limited sensitivity and selectivity and also reported low recovery. Presented is a method for the extraction and HPLC-MS/MS analysis of N-oleoyl glycine and N-oleoyl alanine. The bias and precision of the assay were determined to be within ± 20%. The method was shown to be reliable and robust, with over 90% recovery for the low-level analytes. Increasing concentrations of N-oleoyl glycine and N-oleoyl alanine were quantitated in mouse brain and plasma following exogenous administration. This method was developed to serve to support studies investigating the pharmacokinetics and involvement of N-oleoyl glycine and N-oleoyl alanine in drug dependence and other diseases.


Asunto(s)
Glicina , Espectrometría de Masas en Tándem , Ratones , Animales , Cromatografía Líquida de Alta Presión/métodos , Espectrometría de Masas en Tándem/métodos , Glicina/análisis , Alanina , Encéfalo
5.
J Pharmacol Exp Ther ; 380(1): 1-14, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34625464

RESUMEN

Opioid use disorder reflects a major public health crisis of morbidity and mortality in which opioid withdrawal often contributes to continued use. However, current medications that treat opioid withdrawal symptoms are limited by their abuse liability or lack of efficacy. Although cannabinoid 1 (CB1) receptor agonists, including Δ9-tetrahydrocannabinol, ameliorate opioid withdrawal in both clinical and preclinical studies of opioid dependence, this strategy elicits cannabimimetic side effects as well as tolerance and dependence after repeated administration. Alternatively, CB1 receptor positive allosteric modulators (PAMs) enhance CB1 receptor signaling and show efficacy in rodent models of pain and cannabinoid dependence but lack cannabimimetic side effects. We hypothesize that the CB1 receptor PAM ZCZ011 attenuates naloxone-precipitated withdrawal signs in opioid-dependent mice. Accordingly, male and female mice given an escalating dosing regimen of oxycodone, a widely prescribed opioid, and challenged with naloxone displayed withdrawal signs that included diarrhea, weight loss, jumping, paw flutters, and head shakes. ZCZ011 fully attenuated naloxone-precipitated withdrawal-induced diarrhea and weight loss and reduced paw flutters by approximately half, but its effects on head shakes were unreliable, and it did not affect jumping behavior. The antidiarrheal and anti-weight loss effects of ZCZ0111 were reversed by a CB1 not a cannabinoid receptor type 2 receptor antagonist and were absent in CB1 (-/-) mice, suggesting a necessary role of CB1 receptors. Collectively, these results indicate that ZCZ011 completely blocked naloxone-precipitated diarrhea and weight loss in oxycodone-dependent mice and suggest that CB1 receptor PAMs may offer a novel strategy to treat opioid dependence. SIGNIFICANCE STATEMENT: Opioid use disorder represents a serious public health crisis in which current medications used to treat withdrawal symptoms are limited by abuse liability and side effects. The CB1 receptor positive allosteric modulator (PAM) ZCZ011, which lacks overt cannabimimetic behavioral effects, ameliorated naloxone-precipitated withdrawal signs through a CB1 receptor mechanism of action in a mouse model of oxycodone dependence. These results suggest that CB1 receptor PAMs may represent a viable strategy to treat opioid withdrawal.


Asunto(s)
Antidiarreicos/uso terapéutico , Agonistas de Receptores de Cannabinoides/uso terapéutico , Diarrea/tratamiento farmacológico , Indoles/uso terapéutico , Síndrome de Abstinencia a Sustancias/tratamiento farmacológico , Tiofenos/uso terapéutico , Regulación Alostérica , Animales , Diarrea/etiología , Femenino , Masculino , Ratones , Ratones Endogámicos ICR , Naloxona/efectos adversos , Antagonistas de Narcóticos/efectos adversos , Narcóticos/toxicidad , Trastornos Relacionados con Opioides/tratamiento farmacológico , Trastornos Relacionados con Opioides/etiología , Oxicodona/toxicidad , Receptor Cannabinoide CB1/metabolismo , Síndrome de Abstinencia a Sustancias/etiología
6.
Nat Chem Biol ; 16(6): 667-675, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32393901

RESUMEN

N-acylethanolamines (NAEs), which include the endocannabinoid anandamide, represent an important family of signaling lipids in the brain. The lack of chemical probes that modulate NAE biosynthesis in living systems hamper the understanding of the biological role of these lipids. Using a high-throughput screen, chemical proteomics and targeted lipidomics, we report here the discovery and characterization of LEI-401 as a CNS-active N-acylphosphatidylethanolamine phospholipase D (NAPE-PLD) inhibitor. LEI-401 reduced NAE levels in neuroblastoma cells and in the brain of freely moving mice, but not in NAPE-PLD KO cells and mice, respectively. LEI-401 activated the hypothalamus-pituitary-adrenal axis and impaired fear extinction, thereby emulating the effect of a cannabinoid CB1 receptor antagonist, which could be reversed by a fatty acid amide hydrolase inhibitor. Our findings highlight the distinctive role of NAPE-PLD in NAE biosynthesis in the brain and suggest the presence of an endogenous NAE tone controlling emotional behavior.


Asunto(s)
Conducta Animal/efectos de los fármacos , Inhibidores Enzimáticos/química , Metabolismo de los Lípidos/efectos de los fármacos , Fosfatidiletanolaminas/metabolismo , Fosfolipasa D/antagonistas & inhibidores , Amidohidrolasas/metabolismo , Animales , Proteínas Sanguíneas/metabolismo , Encéfalo/metabolismo , Antagonistas de Receptores de Cannabinoides/metabolismo , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacocinética , Miedo/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Receptores de Cannabinoides/metabolismo , Transducción de Señal
7.
Behav Pharmacol ; 32(5): 422-434, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34050046

RESUMEN

Patients treated with cancer chemotherapeutics frequently report chemotherapy-induced peripheral neuropathy (CIPN), changes in mood (depression and anxiety) and functional impairments. Rodent models of CIPN elicit limited alterations in functional behaviors, which pose challenges in developing preclinical models of chemotherapy-induced behavioral depression. The study examined the consequences of chemotherapy-induced mechanical hypersensitivity (paclitaxel: 32 or 64 mg/kg, cumulative; oxaliplatin: 30 mg/kg, cumulative) on behavioral depression, as measured with operant responding for palatable food during periods of food restriction and ad libitum chow, consumption of noncontingently available palatable food in the presence of ad libitum chow, and voluntary wheel running. The study employed two inbred mouse strains (C57BL/6J and Balb/cJ) and examined potential sex differences. All chemotherapeutic regimens caused profound mechanical hypersensitivity for the duration of the observation periods (up to 7 months), but no treatments changed voluntary wheel running or consumption of noncontingent palatable food. The high dose of paclitaxel temporarily reduced operant responding for palatable food in male C57BL/6J mice undergoing food restriction or maintained on ad libitum chow. However, paclitaxel failed to decrease operant responding for palatable food in free-feeding female C57BL/6J mice or Balb/cJ mice of either sex. Moreover, oxaliplatin did not significantly alter operant responding for palatable food in male or female C57BL/6J mice maintained on ad libitum chow. These findings demonstrate a dissociation between chemotherapy-induced mechanical hypersensitivity and behavioral depression. The transient effects of paclitaxel on operant responding in male C57BL/6J mice may represent a fleeting behavioral correlate of chemotherapy-associated pain-like behaviors.


Asunto(s)
Condicionamiento Operante/efectos de los fármacos , Depresión , Oxaliplatino/farmacología , Paclitaxel/farmacología , Alimentación Animal , Animales , Antineoplásicos/farmacología , Conducta Animal/efectos de los fármacos , Depresión/inducido químicamente , Depresión/psicología , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Dolor/etiología , Dolor/psicología , Factores Sexuales
8.
J Neurosci ; 39(30): 5949-5965, 2019 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-31127001

RESUMEN

Diacylglycerol lipase-α (DAGL-α), the principal biosynthetic enzyme of the endogenous cannabinoid 2-arachidonylglycerol (2-AG) on neurons, plays a key role in CB1 receptor-mediated synaptic plasticity and hippocampal neurogenesis, but its contribution to global hippocampal-mediated processes remains unknown. Thus, the present study examines the role that DAGL-α plays on LTP in hippocampus, as well as in hippocampal-dependent spatial learning and memory tasks, and on the production of endocannabinoid and related lipids through the use of complementary pharmacologic and genetic approaches to disrupt this enzyme in male mice. Here we show that DAGL-α gene deletion or pharmacological inhibition disrupts LTP in CA1 of the hippocampus but elicits varying magnitudes of behavioral learning and memory deficits in mice. In particular, DAGL-α-/- mice display profound impairments in the Object Location assay and Morris Water Maze (MWM) acquisition engaging in nonspatial search strategies. In contrast, WT mice administered the DAGL-α inhibitor DO34 show delays in MWM acquisition and reversal learning, but no deficits in expression, extinction, forgetting, or perseveration processes in this task, as well as no impairment in Object Location. The deficits in synaptic plasticity and MWM performance occur in concert with decreased 2-AG and its major lipid metabolite (arachidonic acid), but increases of a 2-AG diacylglycerol precursor in hippocampus, PFC, striatum, and cerebellum. These novel behavioral and electrophysiological results implicate a direct and perhaps selective role of DAGL-α in the integration of new spatial information.SIGNIFICANCE STATEMENT Here we show that genetic deletion or pharmacologic inhibition of diacylglycerol lipase-α (DAGL-α) impairs hippocampal CA1 LTP, differentially disrupts spatial learning and memory performance in Morris water maze (MWM) and Object Location tasks, and alters brain levels of endocannabinoids and related lipids. Whereas DAGL-α-/- mice exhibit profound phenotypic spatial memory deficits, a DAGL inhibitor selectively impairs the integration of new information in MWM acquisition and reversal tasks, but not memory processes of expression, extinction, forgetting, or perseveration, and does not affect performance in the Objection Location task. The findings that constitutive or short-term DAGL-α disruption impairs learning and memory at electrophysiological and selective in vivo levels implicate this enzyme as playing a key role in the integration of new spatial information.


Asunto(s)
Hipocampo/metabolismo , Lipoproteína Lipasa/metabolismo , Memoria , Aprendizaje Espacial , Animales , Ácido Araquidónico/metabolismo , Hipocampo/fisiología , Lipoproteína Lipasa/antagonistas & inhibidores , Masculino , Ratones , Ratones Endogámicos C57BL
9.
J Neuroinflammation ; 17(1): 345, 2020 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-33208151

RESUMEN

BACKGROUND: Human immunodeficiency virus type-1 (HIV-1) and opiates cause long-term inflammatory insult to the central nervous system (CNS) and worsen disease progression and HIV-1-related neuropathology. The combination of these proinflammatory factors reflects a devastating problem as opioids have high abuse liability and continue to be prescribed for certain patients experiencing HIV-1-related pain. METHODS: Here, we examined the impact of chronic (3-month) HIV-1 transactivator of transcription (Tat) exposure to short-term (8-day), escalating morphine in HIV-1 Tat transgenic mice that express the HIV-1 Tat protein in a GFAP promoter-regulated, doxycycline (DOX)-inducible manner. In addition to assessing morphine-induced tolerance in nociceptive responses organized at spinal (i.e., tail-flick) and supraspinal (i.e., hot-plate) levels, we evaluated neuroinflammation via positron emission tomography (PET) imaging using the [18F]-PBR111 ligand, immunohistochemistry, and cytokine analyses. Further, we examined endocannabinoid (eCB) levels, related non-eCB lipids, and amino acids via mass spectrometry.  RESULTS: Tat-expressing [Tat(+)] transgenic mice displayed antinociceptive tolerance in the tail withdrawal and hot-plate assays compared to control mice lacking Tat [Tat(-)]. This tolerance was accompanied by morphine-dependent increases in Iba-1 ± 3-nitrotryosine immunoreactive microglia, and alterations in pro- and anti-inflammatory cytokines, and chemokines in the spinal cord and striatum, while increases in neuroinflammation were absent by PET imaging of [18F]-PBR111 uptake. Tat and morphine exposure differentially affected eCB levels, non-eCB lipids, and specific amino acids in a region-dependent manner. In the striatum, non-eCB lipids were significantly increased by short-term, escalating morphine exposure, including peroxisome proliferator activator receptor alpha (PPAR-α) ligands N-oleoyl ethanolamide (OEA) and N-palmitoyl ethanolamide (PEA), as well as the amino acids phenylalanine and proline. In the spinal cord, Tat exposure increased amino acids leucine and valine, while morphine decreased levels of tyrosine and valine but did not affect eCBs or non-eCB lipids. CONCLUSION: Overall results demonstrate that 3 months of Tat exposure increased morphine tolerance and potentially innate immune tolerance evidenced by reductions in specific cytokines (e.g., IL-1α, IL-12p40) and microglial reactivity. In contrast, short-term, escalating morphine exposure acted as a secondary stressor revealing an allostatic shift in CNS baseline inflammatory responsiveness from sustained Tat exposure.


Asunto(s)
Aminoácidos/metabolismo , Endocannabinoides/metabolismo , Mediadores de Inflamación/metabolismo , Metabolismo de los Lípidos/fisiología , Morfina/administración & dosificación , Neuroprotección/fisiología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/biosíntesis , Analgésicos Opioides/administración & dosificación , Animales , Relación Dosis-Respuesta a Droga , Mediadores de Inflamación/antagonistas & inhibidores , Metabolismo de los Lípidos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuroprotección/efectos de los fármacos , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
10.
Addict Biol ; 25(1): e12691, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-30378732

RESUMEN

Cessation from prolonged use of ∆9 -tetrahydrocannabinol (THC), the primary active compound responsible for the cannabimimetic effects of cannabis, results in a mild to moderate withdrawal syndrome in humans and laboratory animals. Whereas manipulations of the endogenous cannabinoid system (eg, cannabinoid receptors and endocannabinoid regulating enzymes) alter nicotine withdrawal, in this study we asked the reciprocal question. Do nicotinic acetylcholine receptors (nAChRs) modulate THC withdrawal? To assess the role of different nAChR subtypes in THC withdrawal, we used transgenic mouse, preclinical pharmacological, and human genetic correlation approaches. Our findings show that selective α3ß4* nAChR antagonist, AuIB, and α3ß4* nAChR partial agonist, AT-1001, dose-dependently attenuated somatic withdrawal signs in THC-dependent mice that were challenged with the cannabinoid-1 receptor antagonist rimonabant. Additionally, THC-dependent α5 and α6 nAChR knockout (KO) mice displayed decreased rimonabant precipitated somatic withdrawal signs compared with their wild-type counterparts. In contrast, ß2 and α7 nAChR KO mice showed no alterations in THC withdrawal signs. Moreover, deletion of ß2 nAChR did not alter the reduced expression of somatic signs by the preferred α6ß4* antagonist, BulA [T5A;P60]. Finally, the human genetic association studies indicated that variations in the genes that code for the α5, α3, ß4, and α6 nAChRs were associated with cannabis disorder phenotypes. Overall, these findings suggest that α3ß4* and α6ß4* nAChR subtypes represent viable targets for the development of medications to counteract THC dependence.


Asunto(s)
Dronabinol/farmacología , Abuso de Marihuana/fisiopatología , Receptores Nicotínicos/efectos de los fármacos , Síndrome de Abstinencia a Sustancias/fisiopatología , Animales , Antagonistas de Receptores de Cannabinoides/administración & dosificación , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Rimonabant/administración & dosificación
11.
Handb Exp Pharmacol ; 258: 323-353, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32236882

RESUMEN

Since antiquity, Cannabis has provoked enormous intrigue for its potential medicinal properties as well as for its unique pharmacological effects. The elucidation of its major cannabinoid constituents, Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD), led to the synthesis of new cannabinoids (termed synthetic cannabinoids) to understand the mechanisms underlying the pharmacology of Cannabis. These pharmacological tools were instrumental in the ultimate discovery of the endogenous cannabinoid system, which consists of CB1 and CB2 cannabinoid receptors and endogenously produced ligands (endocannabinoids), which bind and activate both cannabinoid receptors. CB1 receptors mediate the cannabimimetic effects of THC and are highly expressed on presynaptic neurons in the nervous system, where they modulate neurotransmitter release. In contrast, CB2 receptors are primarily expressed on immune cells. The endocannabinoids are tightly regulated by biosynthetic and hydrolytic enzymes. Accordingly, the endocannabinoid system plays a modulatory role in many physiological processes, thereby generating many promising therapeutic targets. An unintended consequence of this research was the emergence of synthetic cannabinoids sold for human consumption to circumvent federal laws banning Cannabis use. Here, we describe research that led to the discovery of the endogenous cannabinoid system and show how knowledge of this system benefitted as well as unintentionally harmed human health.


Asunto(s)
Endocannabinoides/fisiología , Receptor Cannabinoide CB1/fisiología , Receptor Cannabinoide CB2/fisiología , Cannabidiol/farmacología , Cannabinoides/farmacología , Dronabinol/farmacología , Humanos
12.
Drug Metab Rev ; 50(1): 3-13, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29355030

RESUMEN

The cannabinoid receptor 1 (CB1) is a G protein-coupled receptor (GPCR) that is located primarily in the central nervous system. CB1 is a therapeutic target which may impact pathways to mediate pain, neurodegenerative disorders, hunger, and drug-seeking behavior. Despite these benefits, development of orthosteric therapeutic compounds, which target the endogenous ligand-binding site of CB1, has been challenging due to detrimental side effects including psychoactivity, depression, and suicidal thoughts. However, CB1 also has an allosteric binding site(s), which is topographically distinct from the orthosteric site. Allosteric modulation of CB1 has a number of potential advantages including providing a mechanism for more precise control of downstream pathways and circumventing these side effects. In this review, we summarize the concept of allosteric modulation and focus on the structure-activity relationship studies of the well-characterized allosteric modulators, ORG27569 and PSNCBAM-1 and their derivatives, and a few other recent modulators. We review studies on the properties of these modulators on CB1 signaling in cells and their effects in vivo. While many current allosteric modulators also produce complex outcomes, they provide new advances for the design of CB1 centered therapeutics.


Asunto(s)
Indoles/química , Indoles/farmacología , Compuestos de Fenilurea/química , Compuestos de Fenilurea/farmacología , Piperidinas/química , Piperidinas/farmacología , Piridinas/química , Piridinas/farmacología , Receptor Cannabinoide CB1/química , Receptor Cannabinoide CB1/metabolismo , Sitio Alostérico/efectos de los fármacos , Animales , Humanos , Relación Estructura-Actividad , Especificidad por Sustrato
13.
J Pharmacol Exp Ther ; 366(3): 509-518, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29945931

RESUMEN

The immunomodulatory prodrug 2-amino-2-(2-[4-octylphenyl]ethyl)-1,3-propanediol (FTY720), which acts as an agonist for sphingosine-1-phosphate (S1P) receptors (S1PR) when phosphorylated, is proposed as a novel pain therapeutic. In this study, we assessed FTY720-mediated antinociception in the radiant heat tail-flick test and in the chronic constriction injury (CCI) model of neuropathic pain in mice. FTY720 produced antinociception and antiallodynia, respectively, and these effects were dose-dependent and mimicked by the S1PR1-selective agonist CYM-5442. Repeated administration of FTY720 for 1 week produced tolerance to acute thermal antinociception, but not to antiallodynia in the CCI model. S1PR-stimulated [35S]GTPγS autoradiography revealed apparent desensitization of G protein activation by S1P or the S1PR1 agonist 5-[4-phenyl-5-(trifluoromethyl)-2-thienyl]-3-[3-(trifluoromethyl)phenyl]-1,2,4-oxadiazole (SEW-2871) throughout the brain. Similar results were seen in spinal cord membranes, whereby the Emax value of S1PR-stimulated [35S]GTPγS binding was greatly reduced in repeated FTY720-treated mice. These results suggest that S1PR1 is a primary target of FTY720 in alleviating both acute thermal nociception and chronic neuropathic nociception. Furthermore, the finding that tolerance develops to antinociception in the tail-flick test but not in chronic neuropathic pain suggests a differential mechanism of FTY720 action between these models. The observation that repeated FTY720 administration led to desensitized S1PR1 signaling throughout the central nervous system suggests the possibility that S1PR1 activation drives the acute thermal antinociceptive effects, whereas S1PR1 desensitization mediates the following: 1) tolerance to thermal antinociceptive actions of FTY720 and 2) the persistent antiallodynic effects of FTY720 in neuropathic pain by producing functional antagonism of pronociceptive S1PR1 signaling.


Asunto(s)
Clorhidrato de Fingolimod/farmacología , Neuralgia/tratamiento farmacológico , Neuralgia/metabolismo , Péptidos Opioides/efectos de los fármacos , Receptores de Lisoesfingolípidos/metabolismo , Temperatura , Animales , Modelos Animales de Enfermedad , Clorhidrato de Fingolimod/uso terapéutico , Masculino , Ratones , Ratones Endogámicos ICR , Neuralgia/fisiopatología , Receptores de Lisoesfingolípidos/agonistas , Nociceptina
14.
J Pharmacol Exp Ther ; 366(1): 169-183, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29540562

RESUMEN

Although paclitaxel effectively treats various cancers, its debilitating peripheral neuropathic pain side effects often persist long after treatment has ended. Therefore, a compelling need exists for the identification of novel pharmacologic strategies to mitigate this condition. As inhibitors of monoacylglycerol lipase (MAGL), the primary hydrolytic enzyme of the endogenous cannabinoid, 2-arachidonyolglycerol, produces antinociceptive effects in numerous rodent models of pain, we investigated whether inhibitors of this enzyme (i.e., JZL184 and MJN110) would reverse paclitaxel-induced mechanical allodynia in mice. These drugs dose dependently reversed allodynia with respective ED50 values (95% confidence limit) of 8.4 (5.2-13.6) and 1.8 (1.0-3.3) mg/kg. Complementary genetic and pharmacologic approaches revealed that the antiallodynic effects of each drug require both cannabinoid receptors, CB1 and CB2 MJN110 reduced paclitaxel-mediated increased expression of monocyte chemoattractant protein-1 (MCP-1, CCL2) and phospho-p38 MAPK in dorsal root ganglia as well as MCP-1 in spinal dorsal horn. Whereas the antinociceptive effects of high dose JZL184 (40 mg/kg) underwent tolerance following 6 days of repeated dosing, repeated administration of a threshold dose (i.e., 4 mg/kg) completely reversed paclitaxel-induced allodynia. In addition, we found that the administration of MJN110 to control mice lacked intrinsic rewarding effects in the conditioned place preference (CPP) paradigm. However, it produced a CPP in paclitaxel-treated animals, suggesting a reduced paclitaxel-induced aversive state. Importantly, JZL184 did not alter the antiproliferative and apoptotic effects of paclitaxel in A549 and H460 non-small cell lung cancer cells. Taken together, these data indicate that MAGL inhibitors reverse paclitaxel-induced neuropathic pain without interfering with chemotherapeutic efficacy.


Asunto(s)
Antineoplásicos/efectos adversos , Inhibidores Enzimáticos/farmacología , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Monoacilglicerol Lipasas/antagonistas & inhibidores , Nocicepción/efectos de los fármacos , Paclitaxel/efectos adversos , Animales , Apoptosis/efectos de los fármacos , Benzodioxoles/farmacología , Benzodioxoles/uso terapéutico , Biomarcadores/metabolismo , Carbamatos/farmacología , Carbamatos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/uso terapéutico , Humanos , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Inflamación/metabolismo , Masculino , Ratones , Fosfoproteínas/metabolismo , Piperidinas/farmacología , Piperidinas/uso terapéutico , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB2/metabolismo , Succinimidas/farmacología , Succinimidas/uso terapéutico , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
J Pharmacol Exp Ther ; 364(1): 110-119, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29042416

RESUMEN

Chemotherapy-induced peripheral neuropathy (CIPN), a consequence of peripheral nerve fiber dysfunction or degeneration, continues to be a dose-limiting and debilitating side effect during and/or after cancer chemotherapy. Paclitaxel, a taxane commonly used to treat breast, lung, and ovarian cancers, causes CIPN in 59-78% of cancer patients. Novel interventions are needed due to the current lack of effective CIPN treatments. Our studies were designed to investigate whether nicotine can prevent and/or reverse paclitaxel-induced peripheral neuropathy in a mouse model of CIPN, while ensuring that nicotine will not stimulate lung tumor cell proliferation or interfere with the antitumor properties of paclitaxel. Male C57BL/6J mice received paclitaxel every other day for a total of four injections (8 mg/kg, i.p.). Acute (0.3-0.9 mg/kg, i.p.) and chronic (24 mg/kg per day, s.c.) administration of nicotine respectively reversed and prevented paclitaxel-induced mechanical allodynia. Blockade of the antinociceptive effect of nicotine with mecamylamine and methyllycaconitine suggests that the reversal of paclitaxel-induced mechanical allodynia is primarily mediated by the α7 nicotinic acetylcholine receptor subtype. Chronic nicotine treatment also prevented paclitaxel-induced intraepidermal nerve fiber loss. Notably, nicotine neither promoted proliferation of A549 and H460 non-small cell lung cancer cells nor interfered with paclitaxel-induced antitumor effects, including apoptosis. Most importantly, chronic nicotine administration did not enhance Lewis lung carcinoma tumor growth in C57BL/6J mice. These data suggest that the nicotinic acetylcholine receptor-mediated pathways may be promising drug targets for the prevention and treatment of CIPN.


Asunto(s)
Hiperalgesia/tratamiento farmacológico , Hiperalgesia/prevención & control , Nicotina/farmacología , Paclitaxel/efectos adversos , Animales , Antineoplásicos Fitogénicos/farmacología , Hidrocarburos Aromáticos con Puentes/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Modelos Animales de Enfermedad , Hiperalgesia/inducido químicamente , Neoplasias Pulmonares/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Paclitaxel/farmacología , Enfermedades del Sistema Nervioso Periférico/inducido químicamente , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Enfermedades del Sistema Nervioso Periférico/prevención & control , Receptores Colinérgicos/metabolismo , Taxoides/farmacología
16.
Mol Pharm ; 15(3): 721-728, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-28901776

RESUMEN

Diacylglycerol lipase-beta (DAGLß) hydrolyzes arachidonic acid (AA)-containing diacylglycerols to produce bioactive lipids including endocannabinoids and AA-derived eicosanoids involved in regulation of inflammatory signaling. Previously, we demonstrated that DAGLß inactivation using the triazole urea inhibitor KT109 blocked macrophage inflammatory signaling and reversed allodynic responses of mice in inflammatory and neuropathic pain models. Here, we tested whether we could exploit the phagocytic capacity of macrophages to localize delivery of DAGLß inhibitors to these cells in vivo using liposome encapsulated KT109. We used DAGLß-tailored activity-based probes and chemical proteomic methods to measure potency and selectivity of liposomal KT109 in macrophages and tissues from treated mice. Surprisingly, delivery of ∼5 µg of liposomal KT109 was sufficient to achieve ∼80% inactivation of DAGLß in macrophages with no apparent activity in other tissues in vivo. Our macrophage-targeted delivery resulted in a >100-fold enhancement in antinociceptive potency compared with free compound in a mouse inflammatory pain model. Our studies describe a novel anti-inflammatory strategy that is achieved by targeted in vivo delivery of DAGLß inhibitors to macrophages.


Asunto(s)
Antiinflamatorios/farmacología , Inhibidores Enzimáticos/farmacología , Lipoproteína Lipasa/antagonistas & inhibidores , Macrófagos/efectos de los fármacos , Dolor/tratamiento farmacológico , Fagocitosis/efectos de los fármacos , Triazoles/farmacología , Urea/análogos & derivados , Urea/farmacología , Animales , Antiinflamatorios/uso terapéutico , Conducta Animal/efectos de los fármacos , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/uso terapéutico , Humanos , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/etiología , Inflamación/complicaciones , Inflamación/inmunología , Lipopolisacáridos/inmunología , Lipoproteína Lipasa/metabolismo , Liposomas , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Dolor/inmunología , Fagocitosis/inmunología , Resultado del Tratamiento , Triazoles/uso terapéutico , Urea/uso terapéutico
17.
Mol Cell Neurosci ; 83: 92-102, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28733129

RESUMEN

In the era of combined antiretroviral therapy (cART), human immunodeficiency virus type 1 (HIV-1) is considered a chronic disease that specifically targets the brain and causes HIV-1-associated neurocognitive disorders (HAND). Endocannabinoids (eCBs) elicit neuroprotective and anti-inflammatory actions in several central nervous system (CNS) disease models, but their effects in HAND remain unknown. HIV-1 does not infect neurons, but produces viral toxins, such as transactivator of transcription (Tat), that disrupt neuronal calcium equilibrium and give rise to synaptodendritic injuries and cell death, the former being highly correlated with HAND. Consequently, we tested whether the eCBs N-arachidonoylethanolamine (anandamide/AEA) and 2-arachidonoyl-glycerol (2-AG) offer neuroprotective actions in a neuronal culture model. Specifically, we examined the neuroprotective actions of these eCBs on Tat excitotoxicity in primary cultures of prefrontal cortex neurons (PFC), and whether cannabinoid receptors mediate this neuroprotection. Tat-induced excitotoxicity was reflected by increased intracellular calcium levels, synaptodendritic damage, neuronal excitability, and neuronal death. Further, upregulation of cannabinoid 1 receptor (CB1R) protein levels was noted in the presence of HIV-1 Tat. The direct application of AEA and 2-AG reduced excitotoxic levels of intracellular calcium and promoted neuronal survival following Tat exposure, which was prevented by the CB1R antagonist rimonabant, but not by the CB2R antagonist AM630. Overall, our findings indicate that eCBs protect PFC neurons from Tat excitotoxicity in vitro via a CB1R-related mechanism. Thus, the eCB system possesses promising targets for treatment of neurodegenerative disorders associated with HIV-1 infection.


Asunto(s)
Ácidos Araquidónicos/farmacología , Endocannabinoides/farmacología , Glicéridos/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Receptor Cannabinoide CB1/agonistas , Animales , Calcio/metabolismo , Antagonistas de Receptores de Cannabinoides/farmacología , Supervivencia Celular , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Neuronas/fisiología , Piperidinas/farmacología , Alcamidas Poliinsaturadas , Corteza Prefrontal/citología , Pirazoles/farmacología , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB1/metabolismo , Rimonabant , Transmisión Sináptica , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/toxicidad
18.
J Pharmacol Exp Ther ; 363(3): 394-401, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28970359

RESUMEN

Diacylglycerol lipase (DAGL) α and ß, the major biosynthetic enzymes of the endogenous cannabinoid (endocannabinoid) 2-arachidonylglycerol (2-AG), are highly expressed in the nervous system and immune system, respectively. Genetic deletion or pharmacological inhibition of DAGL-ß protects against lipopolysaccharide (LPS)-induced inflammatory responses in mouse peritoneal macrophages and reverses LPS-induced allodynia in mice. To gain insight into the contribution of DAGL-α in LPS-induced allodynia, we tested global knockout mice as well as DO34, a dual DAGL-α/ß inhibitor. Intraperitoneal administration of DO34 (30 mg/kg) significantly decreased whole-brain levels of 2-AG (∼83%), anandamide (∼42%), and arachidonic acid (∼58%). DO34 dose-dependently reversed mechanical and cold allodynia, and these antinociceptive effects did not undergo tolerance after 6 days of repeated administration. In contrast, DO34 lacked acute thermal antinociceptive, motor, and hypothermal pharmacological effects in naive mice. As previously reported, DAGL-ß (-/-) mice displayed a protective phenotype from LPS-induced allodynia. However, DAGL-α (-/-) mice showed full allodynic responses, similar to their wild-type littermates. Interestingly, DO34 (30 mg/kg) fully reversed LPS-induced allodynia in DAGL-α (+/+) and (-/-) mice, but did not affect the antinociceptive phenotype of DAGL-ß (-/-) mice in this model, indicating a DAGL-α-independent site of action. These findings suggest that DAGL-α and DAGL-ß play distinct roles in LPS-induced nociception. Whereas DAGL-α appears to be dispensable for the development and expression of LPS-induced nociception, DAGL-ß inhibition represents a promising strategy to treat inflammatory pain.


Asunto(s)
Analgésicos/farmacología , Lipopolisacáridos/farmacología , Lipoproteína Lipasa/antagonistas & inhibidores , Dolor/enzimología , Tiazoles/farmacología , Urea/análogos & derivados , Urea/farmacología , Analgésicos/uso terapéutico , Animales , Temperatura Corporal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Tolerancia a Medicamentos , Endocannabinoides/metabolismo , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/fisiopatología , Inflamación/tratamiento farmacológico , Inflamación/enzimología , Inflamación/fisiopatología , Inflamación/psicología , Lipoproteína Lipasa/genética , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Nocicepción/efectos de los fármacos , Dolor/tratamiento farmacológico , Dolor/fisiopatología , Dolor/psicología , Tiazoles/uso terapéutico , Urea/uso terapéutico
19.
Neurobiol Dis ; 92(Pt B): 124-36, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26845176

RESUMEN

Co-exposure to opiates and HIV/HIV proteins results in enhanced CNS morphological and behavioral deficits in HIV(+) individuals and in animal models. Opiates with abuse liability, such as heroin and morphine, bind preferentially to and have pharmacological actions through µ-opioid-receptors (MORs). The mechanisms underlying opiate-HIV interactions are not understood. Exposure to the HIV-1 transactivator of transcription (Tat) protein causes neurodegenerative outcomes that parallel many aspects of the human disease. We have also observed that in vivo exposure to Tat results in apparent changes in morphine efficacy, and thus have hypothesized that HIV proteins might alter MOR activation. To test our hypothesis, MOR-mediated G-protein activation was determined in neuroAIDS-relevant forebrain regions of transgenic mice with inducible CNS expression of HIV-1 Tat. G-protein activation was assessed by MOR agonist-stimulated [(35)S]guanosine-5'-O-(3-thio)triphosphate ([(35)S]GTPγS) autoradiography in brain sections, and in concentration-effect curves of MOR agonist-stimulated [(35)S]GTPγS binding in membranes isolated from specific brain regions. Comparative studies were done using the MOR-selective agonist DAMGO ([D-Ala(2), N-MePhe(4), Gly-ol]-enkephalin) and a more clinically relevant agonist, morphine. Tat exposure reduced MOR-mediated G-protein activation in an agonist, time, and regionally dependent manner. Levels of the GPCR regulatory protein ß-arrestin-2, which is involved in MOR desensitization, were found to be elevated in only one affected brain region, the amygdala; amygdalar ß-arrestin-2 also showed a significantly increased association with MOR by co-immunoprecipitation, suggesting decreased availability of MOR. Interestingly, this correlated with changes in anxiety and fear-conditioned extinction, behaviors that have substantial amygdalar input. We propose that HIV-1 Tat alters the intrinsic capacity of MOR to signal in response to agonist binding, possibly via a mechanism involving altered expression and/or function of ß-arrestin-2.


Asunto(s)
Ansiedad/metabolismo , Miedo/fisiología , Prosencéfalo/metabolismo , Receptores Opioides mu/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Complejo SIDA Demencia/metabolismo , Analgésicos Opioides/farmacología , Animales , Ansiedad/virología , Condicionamiento Psicológico/fisiología , Encefalina Ala(2)-MeFe(4)-Gli(5)/farmacología , Proteínas de Unión al GTP/metabolismo , VIH-1 , Masculino , Ratones , Ratones Transgénicos , Morfina/farmacología , Prosencéfalo/efectos de los fármacos , Receptores Opioides mu/agonistas , Arrestina beta 2/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética
20.
J Pharmacol Exp Ther ; 359(2): 310-318, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27608657

RESUMEN

Chemotherapy-induced peripheral neuropathy (CIPN) represents a serious complication associated with antineoplastic drugs. Although there are no medications available that effectively prevent CIPN, many classes of drugs have been used to treat this condition, including anticonvulsants, serotonin and noradrenaline reuptake inhibitors, and opioids. However, these therapeutic options yielded inconclusive results in CIPN clinical trials and produced assorted side effects with their prolonged use. Thus, there is an urgent need to develop efficacious and safe treatments for CIPN. In this report, we tested whether the endogenous lipid palmitoylethanolamide (PEA) alone or in combination with the anticonvulsant gabapentin would reduce allodynia in a mouse paclitaxel model of CIPN. Gabapentin and PEA reversed paclitaxel-induced allodynia with respective ED50 doses (95% confidence interval) of 67.4 (61.52-73.94) and 9.2 (8.39-10.16) mg/kg. Isobolographic analysis of these drugs in combination revealed synergistic antiallodynic effects. The PPAR-α antagonist receptor antagonist GW6471 [N-((2S)-2-(((1Z)-1-methyl-3-oxo-3-(4-(trifluoromethyl)phenyl)prop-1-enyl)amino)-3-(4-(2-(5-methyl-2-phenyl-1,3-oxazol-4-yl)ethoxy)phenyl)propyl)propanamide] completely blocked the antinociceptive effects of PEA. In addition, PEA administered via intraplantar injection into a paw, intrathecal injection, and intracerebroventricular injection reversed paclitaxel-induced allodynia, suggesting that it may act at multiple sites in the neuroaxis and periphery. Finally, repeated administration of PEA (30 mg/kg, 7 days) preserved the antiallodynic effects with no evidence of tolerance. These findings taken together suggest that PEA possesses potential to treat peripheral neuropathy in cancer patients undergoing chemotherapy.


Asunto(s)
Etanolaminas/farmacología , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Paclitaxel/efectos adversos , Ácidos Palmíticos/farmacología , Amidas , Aminas/farmacología , Animales , Ácidos Ciclohexanocarboxílicos/farmacología , Sinergismo Farmacológico , Etanolaminas/administración & dosificación , Etanolaminas/uso terapéutico , Gabapentina , Hiperalgesia/metabolismo , Masculino , Ratones , PPAR alfa/metabolismo , Ácidos Palmíticos/administración & dosificación , Ácidos Palmíticos/uso terapéutico , Ácido gamma-Aminobutírico/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA