Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
1.
Eur J Med Chem ; 228: 113986, 2022 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-34802839

RESUMEN

Biased agonism refers to the ability of compounds to drive preferred signaling pathways and avoid adverse signaling pathways in a ligand-dependent manner for some G-protein-coupled receptors. It is thought that the separation of therapeutic efficacy (e.g., analgesia) from adverse effects (e.g., respiration depression) can be achieved through the design of biased MOR agonists and one example is the recently approved MOR biased agonist oliceridine (TRV130). However, oliceridine only demonstrates modest beneficial effects as compared to other opioids in terms of therapeutic/adverse effect balance. One possibility attributable to the modest success of oliceridine is its limited bias, and as such developing MOR ligands with a more biased agonism profile could in theory further improve the beneficial effects of the ligands. Here, we rationally designed and synthesized a series of derivatives as potent highly biased MOR agonists (19a-v) through the modification and structure-activity relationship study of TRV130. This novel synthetic molecule, LPM3480392 (19m), demonstrated improved in vitro biased agonism (EC50 = 0.35 nM, Emax = 91.4%) with no measured ß-arrestin recruitment (EC50 > 30000 nM, Emax = 1.6%), good brain penetration (B/P ratio = 4.61, 0.25 h post-IV dosing 2.0 mg/kg), a favorable pharmacokinetic profile (distribution volume = 10766 mL/kg, t1/2 = 1.9 h) and produced potent antinociceptive effect with reduced respiratory suppression (sO2(%) = 92.17, 0.32 mg/kg, SC) as compared to TRV130. LPM3480392 has completed preclinical studies and is currently under clinical development (CTR20210370) as an analgesic for the treatment of moderate to severe pain.


Asunto(s)
Analgésicos Opioides/farmacología , Proteínas de Unión al GTP/agonistas , Receptores Opioides mu/agonistas , Analgésicos Opioides/química , Animales , Perros , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Microsomas Hepáticos/química , Microsomas Hepáticos/metabolismo , Estructura Molecular , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
2.
Am J Physiol Cell Physiol ; 321(4): C681-C683, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34469203

RESUMEN

Respiratory depression is a potentially fatal side effect of opioid analgesics and a major limitation to their use. G protein-biased opioid agonists have been proposed as "safer" analgesics with less respiratory depression. These agonists are biased to activate G proteins rather than ß-arrestin signaling. Respiratory depression has been shown to correlate with both G protein bias and intrinsic efficacy, and recent work has refuted the role of ß-arrestin signaling in opioid-induced respiratory depression. In addition, there is substantial evidence that G proteins do, in fact, mediate respiratory depression by actions in respiratory-controlling brainstem neurons. Based on these studies, we provide the perspective that protection from respiratory depression displayed by newly developed G protein-biased agonists is due to factors other than G protein versus arrestin bias.


Asunto(s)
Analgésicos Opioides/efectos adversos , Proteínas de Unión al GTP/agonistas , Pulmón/efectos de los fármacos , Respiración/efectos de los fármacos , Insuficiencia Respiratoria/inducido químicamente , Arrestina beta 2/metabolismo , Animales , Proteínas de Unión al GTP/metabolismo , Humanos , Pulmón/metabolismo , Pulmón/fisiopatología , Insuficiencia Respiratoria/metabolismo , Insuficiencia Respiratoria/fisiopatología , Insuficiencia Respiratoria/prevención & control , Factores de Riesgo , Transducción de Señal
3.
J Mol Endocrinol ; 67(2): 41-53, 2021 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-34077389

RESUMEN

The calcium-sensing receptor (CaSR) is a G protein-coupled receptor that plays a fundamental role in extracellular calcium (Ca2+e) homeostasis by regulating parathyroid hormone release and urinary calcium excretion. The CaSR has been described to activate all four G protein subfamilies (Gαq/11, Gαi/o, Gα12/13, Gαs), and mutations in the receptor that cause hyper/hypocalcaemia, have been described to bias receptor signalling. However, many of these studies are based on measurements of second messengers or gene transcription that occurs many steps downstream of receptor activation and can represent convergence points of several signalling pathways. Therefore, to assess CaSR-mediated G protein activation directly, we took advantage of a recently described NanoBiT G protein dissociation assay system. Our studies, performed in HEK293 cells stably expressing CaSR, demonstrate that Ca2+e stimulation activates all Gαq/11 family and several Gαi/o family proteins, although Gαz was not activated. CaSR stimulated dissociation of Gα12/13 and Gαs from Gß-subunits, but this occurred at a slower rate than that of other Gα-subunits. Investigation of cDNA expression of G proteins in three tissues abundantly expressing CaSR, the parathyroids, kidneys and pancreas, showed Gα11, Gαz, Gαi1 and Gα13 genes were highly expressed in parathyroid tissue, indicating CaSR most likely activates Gα11 and Gαi1 in parathyroids. In kidney and pancreas, the majority of G proteins were similarly expressed, suggesting CaSR may activate multiple G proteins in these cells. Thus, these studies validate a single assay system that can be used to robustly assess CaSR variants and biased signalling and could be utilised in the development of new pharmacological compounds targeting CaSR.


Asunto(s)
Proteínas de Unión al GTP/agonistas , Receptores Sensibles al Calcio/metabolismo , Biomarcadores , AMP Cíclico/metabolismo , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Riñón/metabolismo , Familia de Multigenes , Especificidad de Órganos/genética , Páncreas/metabolismo , Glándulas Paratiroides/metabolismo , Unión Proteica , Isoformas de Proteínas , Receptores Sensibles al Calcio/genética
4.
Angew Chem Int Ed Engl ; 60(18): 10247-10254, 2021 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-33596327

RESUMEN

G protein-coupled receptors (GPCRs) represent an important group of membrane proteins that play a central role in modern medicine. Unfortunately, conformational promiscuity hampers full therapeutic exploitation of GPCRs, since the largest population of the receptor will adopt a basal conformation, which subsequently challenges screens for agonist drug discovery programs. Herein, we describe a set of peptidomimetics able to mimic the ability of G proteins in stabilizing the active state of the ß2 adrenergic receptor (ß2 AR) and the dopamine 1 receptor (D1R). During fragment-based screening efforts, these (un)constrained peptide analogues of the α5 helix in Gs proteins, were able to identify agonism pre-imprinted fragments for the examined GPCRs, and as such, they behave as a generic tool, enabling an engagement in agonist earmarked discovery programs.


Asunto(s)
Descubrimiento de Drogas , Proteínas de Unión al GTP/agonistas , Peptidomiméticos/farmacología , Receptores Acoplados a Proteínas G/agonistas , Proteínas de Unión al GTP/metabolismo , Humanos , Modelos Moleculares , Peptidomiméticos/síntesis química , Peptidomiméticos/química , Receptores Acoplados a Proteínas G/metabolismo
5.
Cell Signal ; 80: 109906, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33383156

RESUMEN

Opioid analgesics are elective for treating moderate to severe pain but their use is restricted by severe side effects. Signaling bias has been proposed as a viable means for improving this situation. To exploit this opportunity, continuous efforts are devoted to understand how ligand-specific modulations of receptor functions could mediate the different in vivo effects of opioids. Advances in the field have led to the development of biased agonists based on hypotheses that allocated desired and undesired effects to specific signaling pathways. However, the prevalent hypothesis associating ß-arrestin to opioid side effects was recently challenged and multiple of the newly developed biased drugs may not display the superior side effects profile that was sought. Moreover, biased agonism at opioid receptors is now known to be time- and cell-dependent, which adds a new layer of complexity for bias estimation. Here, we first review the signaling mechanisms underlying desired and undesired effects of opioids. We then describe biased agonism at opioid receptors and discuss the different perspectives that support the desired and undesired effects of opioids in view of exploiting biased signaling for therapeutic purposes. Finally, we explore how signaling kinetics and cellular background can influence the magnitude and directionality of bias at those receptors.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Transducción de Señal , beta-Arrestinas/metabolismo , Analgésicos Opioides/farmacología , Proteínas de Unión al GTP/agonistas , Humanos , Cinética , Ligandos , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Transducción de Señal/efectos de los fármacos , beta-Arrestinas/agonistas
6.
Eur J Pharmacol ; 890: 173640, 2021 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-33045198

RESUMEN

Opioids are the most powerful analgesics used clinically; however, severe side effects limit their long-term use. Various concepts involving biased intracellular signaling, partial agonism or multi-receptor targeting have been proposed to identify novel opioids with increased analgesic efficacy but reduced side effects. The search for such 'better opioids' implies screening of huge compound libraries and requires highly reliable, easy to perform and high throughput screening (HTS) assays. Here, we utilize an established membrane potential assay to monitor activation of G protein-coupled inwardly rectifying potassium (GIRK) channels, one of the main effectors of opioid receptor signaling, as readout to determine pharmacological profiles of opioids in a non-invasive manner. Specifically, in this study, we optimize assay conditions and extend the application of this assay to screen all four members of the opioid receptor family, stably expressed in AtT-20 and HEK293 cells. This ultra-sensitive system yielded EC50 values in the nano-molar range. We further validate this system for screening cells stably co-expressing two opioid receptors, which could be a valuable tool for investigating bi-functional ligands and studying interactions between receptors. Additionally, we demonstrate the utility of this assay to study antagonists as well as ligands with varying efficacies. Our results suggest that this assay could easily be up-scaled to HTS assay in order to efficiently study receptor activation and screen for novel opioids.


Asunto(s)
Proteínas de Unión al GTP/efectos de los fármacos , Proteínas de Unión al GTP/metabolismo , Ensayos Analíticos de Alto Rendimiento/métodos , Potenciales de la Membrana/efectos de los fármacos , Receptores Opioides/metabolismo , Transducción de Señal/efectos de los fármacos , Analgésicos Opioides/farmacología , Animales , Línea Celular Tumoral , Separación Celular , Citometría de Flujo , Fluorescencia , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/agonistas , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/antagonistas & inhibidores , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/efectos de los fármacos , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Proteínas de Unión al GTP/agonistas , Proteínas de Unión al GTP/antagonistas & inhibidores , Células HEK293 , Humanos , Ligandos , Ratones
7.
PLoS One ; 15(10): e0241192, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33095824

RESUMEN

Mammary-derived serotonin has been implicated in breast-to-bone communication during lactation by increasing parathyroid hormone related-protein (PTHrP) in the mammary gland. It is well established that PTHrP acts on the bone to liberate calcium for milk synthesis during lactation; however, the mechanism of serotonin's regulation of PTHrP has not been fully elucidated. Recently, serotonylation has been shown to be involved in a variety of physiological processes mediated by serotonin. Therefore, we investigated whether serotonylation is involved in serotonin's regulation of PTHrP in the mammary gland using lactogenically differentiated mouse mammary epithelial cells. We investigated the effect of increased intracellular serotonin using the antidepressant fluoxetine or 5-hydroxytryptophan (serotonin precursor), with or without transglutaminase inhibition and the corresponding action on PTHrP induction and activity. Treatment with fluoxetine or 5-hydroxytryptophan significantly increased intracellular serotonin concentrations and subsequently increased PTHrP gene expression, which was reduced with transglutaminase inhibition. Furthermore, we determined that transglutaminase activity is increased with lactogenic differentiation and 5-hydroxytryptophan or fluoxetine treatment. We investigated whether RhoA, Rac1, and Rab4 were potential serotonylation target proteins. We speculate that RhoA is potentially a serotonylation target protein. Our data suggest that serotonin regulates PTHrP induction in part through the process of serotonylation under lactogenic conditions in mouse mammary epithelial cells.


Asunto(s)
Células Epiteliales/metabolismo , Proteínas de Unión al GTP/metabolismo , Glándulas Mamarias Animales/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Serotonina/metabolismo , Transglutaminasas/metabolismo , 5-Hidroxitriptófano/farmacología , Animales , Huesos/metabolismo , Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular , Células Epiteliales/efectos de los fármacos , Femenino , Fluoxetina/farmacología , Proteínas de Unión al GTP/agonistas , Lactancia/efectos de los fármacos , Lactancia/metabolismo , Glándulas Mamarias Animales/citología , Ratones , Prolactina/farmacología , Proteína Glutamina Gamma Glutamiltransferasa 2 , Regulación hacia Arriba/efectos de los fármacos , Proteína de Unión al GTP rhoA/metabolismo
8.
Pharmacol Res ; 160: 105148, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32858121

RESUMEN

Neuropathic pain, a specific type of chronic pain resulting from persistent nervous tissue lesions, is a debilitating condition that affects about 7% of the population. This condition remains particularly difficult to treat because of the poor understanding of its underlying mechanisms. Drugs currently used to alleviate this chronic pain syndrome are of limited benefit due to their lack of efficacy and the elevated risk of side effects, especially after a prolonged period of treatment. Although drugs targeting G protein-coupled receptors (GPCR) also have several limitations, such as progressive loss of efficacy due to receptor desensitization or unavoidable side effects due to wide receptor distribution, the identification of several molecular partners that contribute to the fine-tuning of receptor activity has raised new opportunities for the development of alternative therapeutic approaches. Regulators of G protein signalling (RGS) act intracellularly by influencing the coupling process and activity of G proteins, and are amongst the best-characterized physiological modulators of GPCR. Changes in RGS expression have been documented in a range of models of neuropathic pain, or after prolonged treatment with diverse analgesics, and could participate in altered pain processing as well as impaired physiological or pharmacological control of nociceptive signals. The present review summarizes the experimental data that implicates RGS in the development of pain with focus on the pathological mechanisms of neuropathic pain, including the impact of neuropathic lesions on RGS expression and, reciprocally, the influence of modifying RGS on GPCRs involved in the modulation of nociception as well as on the outcome of pain. In this context, we address the question of the relevance of RGS as promising targets in the treatment of neuropathic pain.


Asunto(s)
Proteínas de Unión al GTP/efectos de los fármacos , Neuralgia/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Animales , Dolor Crónico , Proteínas de Unión al GTP/agonistas , Humanos , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/efectos de los fármacos
9.
Artículo en Inglés | MEDLINE | ID: mdl-32676053

RESUMEN

G protein coupled receptors (GPCRs) can lead to G protein and non-G protein initiated signals. By virtue of its structural property, the TSH receptor (TSHR) has a unique ability to engage different G proteins making it highly amenable to selective signaling. In this study, we describe the identification and characterization of a novel small molecule agonist to the TSHR which induces primary engagement with Gαq/11. To identify allosteric modulators inducing selective signaling of the TSHR we used a transcriptional-based luciferase assay system with CHO-TSHR cells stably expressing response elements (CRE, NFAT, SRF, or SRE) that were capable of measuring signals emanating from the coupling of Gαs , Gαq/11, Gßγ, and Gα12/13, respectively. Using this system, TSH activated Gαs , Gαq/11, and Gα12/13 but not Gßγ. On screening a library of 50K molecules at 0.1,1.0 and 10 µM, we identified a novel Gq/11 agonist (named MSq1) which activated Gq/11 mediated NFAT-luciferase >4 fold above baseline and had an EC50= 8.3 × 10-9 M with only minor induction of Gαs and cAMP. Furthermore, MSq1 is chemically and structurally distinct from any of the previously reported TSHR agonist molecules. Docking studies using a TSHR transmembrane domain (TMD) model indicated that MSq1 had contact points on helices H1, H2, H3, and H7 in the hydrophobic pocket of the TMD and also with the extracellular loops. On co-treatment with TSH, MSq1 suppressed TSH-induced proliferation of thyrocytes in a dose-dependent manner but lacked the intrinsic ability to influence basal thyrocyte proliferation. This unexpected inhibitory property of MSq1 could be blocked in the presence of a PKC inhibitor resulting in derepressing TSH induced protein kinase A (PKA) signals and resulting in the induction of proliferation. Thus, the inhibitory effect of MSq1 on proliferation resided in its capacity to overtly activate protein kinase C (PKC) which in turn suppressed the proliferative signal induced by activation of the predomiant cAMP-PKA pathway of the TSHR. Treatment of rat thyroid cells (FRTL5) with MSq1 did not show any upregulation of gene expression of the key thyroid specific markers such as thyroglobulin(Tg), thyroid peroxidase (Tpo), sodium iodide symporter (Nis), and the TSH receptor (Tshr) further suggesting lack of involvement of MSq1 and Gαq/11 activation with cellular differentation. In summary, we identified and characterized a novel Gαq/11 agonist molecule acting at the TSHR and which showed a marked anti-proliferative ability. Hence, Gq biased activation of the TSHR is capable of ameliorating the proliferative signals from its orthosteric ligand and may offer a therapeutic option for thyroid growth modulation.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Receptores de Tirotropina/metabolismo , Transducción de Señal , Animales , Células CHO , Proliferación Celular , Cricetulus , Proteínas de Unión al GTP/agonistas , Simulación del Acoplamiento Molecular , Unión Proteica , Células Epiteliales Tiroideas/metabolismo
10.
Drugs Today (Barc) ; 56(4): 269-286, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32309822

RESUMEN

Oliceridine is a next-generation investigational intravenous opioid that is a G protein-selective agonist at the µ-opioid receptor. The G protein selectivity of this compound results in potent analgesia with substantially reduced recruitment of ß-arrestin, a signaling pathway associated with opioid-related adverse events. In randomized, placebo- and active-controlled clinical studies, use of oliceridine for the management of moderate to severe acute pain provided potent analgesic effect superior to that observed with placebo, with lower incidence of adverse events, including respiratory events and gastrointestinal events of nausea and vomiting, compared with morphine. Here, we provide a review of the preclinical and clinical data of intravenous oliceridine, a selective agonist, which has the potential to offer a wider therapeutic window than conventional opioids.


Asunto(s)
Dolor Agudo/tratamiento farmacológico , Analgésicos Opioides/uso terapéutico , Compuestos de Espiro/uso terapéutico , Tiofenos/uso terapéutico , Proteínas de Unión al GTP/agonistas , Humanos , Morfina , Ensayos Clínicos Controlados Aleatorios como Asunto , Receptores Opioides mu
11.
Biochem Pharmacol ; 177: 113910, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32179045

RESUMEN

Fentanyl and morphine are agonists of the Mu opioid receptor (MOR), which is a member of the GPCR family. Their analgesic effects are associated with unwanted side effects. On a signaling level downstream from MOR, it has been hypothesized that analgesia may be mediated through the G protein pathway, whereas the undesirable effects of opioids have been linked to the ß-arrestin (ßarr) pathway. Despite being an increasingly debated subject, little is known about a potential 'bias' (i.e. the preferential activation of one pathway over the other) of the novel synthetic opioids (NSO) - including fentanyl analogs - that have emerged on the illegal drug market. We have therefore developed and applied a novel, robust bio-assay platform to study the activity of 21 NSO, to evaluate to what extent these MOR agonists show biased agonism and to investigate the potential correlation with their structure. In addition, we evaluated the functional selectivity of TRV130, a purported G protein-biased agonist. We applied newly established stable bio-assays in HEK293T cells, based on the principle of functional complementation of a split nanoluciferase, to assess MOR activation via recruitment of a mini-Gi protein (GTPase domain of Gαi subunit) or ßarr2. All but two of the tested NSO demonstrated a concentration-dependent response at MOR in both bio-assays. The developed bio-assays allow to gain insight into the ßarr2 or G protein recruitment potential of NSO, which may eventually help to better understand why certain opioids are associated with higher toxicity. Adding to the recent discussion about the relevance of the biased agonism concept for opioids, we did not observe a significant bias for any of the evaluated compounds, including TRV130.


Asunto(s)
Analgésicos Opioides/química , Analgésicos Opioides/farmacología , Bioensayo/métodos , Receptores Opioides mu/agonistas , Transducción de Señal/efectos de los fármacos , Drogas Sintéticas/química , Drogas Sintéticas/farmacología , Citometría de Flujo/métodos , Proteínas de Unión al GTP/agonistas , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Humanos , Mapas de Interacción de Proteínas , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Compuestos de Espiro/química , Compuestos de Espiro/farmacología , Relación Estructura-Actividad , Tiofenos/química , Tiofenos/farmacología , Transducción Genética/métodos , Transfección/métodos , Arrestina beta 2/genética , Arrestina beta 2/metabolismo
12.
Sci Rep ; 9(1): 19295, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31848378

RESUMEN

The neuropeptides oxytocin (OT) and vasopressin (VP) and their G protein-coupled receptors OTR, V1aR, V1bR, and V2R form an important and widely-distributed neuroendocrine signaling system. In mammals, this signaling system regulates water homeostasis, blood pressure, reproduction, as well as social behaviors such as pair bonding, trust and aggression. There exists high demand for ligands with differing pharmacological profiles to study the physiological and pathological functions of the individual receptor subtypes. Here, we present the pharmacological characterization of an arthropod (Metaseiulus occidentalis) OT/VP-like nonapeptide across the human OT/VP receptors. I8-arachnotocin is a full agonist with respect to second messenger signaling at human V2R (EC50 34 nM) and V1bR (EC50 1.2 µM), a partial agonist at OTR (EC50 790 nM), and a competitive antagonist at V1aR [pA2 6.25 (558 nM)]. Intriguingly, I8-arachnotocin activated the Gαs pathway of V2R without recruiting either ß-arrestin-1 or ß-arrestin-2. I8-arachnotocin might thus be a novel pharmacological tool to study the (patho)physiological relevance of ß-arrestin-1 or -2 recruitment to the V2R. These findings furthermore highlight arthropods as a novel, vast and untapped source for the discovery of novel pharmacological probes and potential drug leads targeting neurohormone receptors.


Asunto(s)
Artrópodos/química , Neuropéptidos/agonistas , Receptores de Vasopresinas/agonistas , Vasopresinas/agonistas , Animales , Proteínas de Unión al GTP/agonistas , Humanos , Ligandos , Neuropéptidos/química , Neuropéptidos/farmacología , Oxitocina/agonistas , Oxitocina/química , Oxitocina/farmacología , Unión Proteica/efectos de los fármacos , Receptores Acoplados a Proteínas G/genética , Receptores de Vasopresinas/química , Transducción de Señal/genética , Vasopresinas/química
13.
Neuroscience ; 404: 246-258, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30794845

RESUMEN

Bidirectional selection of mice for high (HA) and low (LA) swim stress-induced analgesia (SSIA) is associated with a divergent response to opioids. In the current study, we investigated whether the genetic divergence in opioid system activity between HA and LA mice also affects cannabinoid sensitivity. Additionally, we also investigated whether the endocannabinoid system mediates SSIA in these lines. Numerous reports support the existence of pharmacological and molecular interactions between the opioid and cannabinoid systems along the pain pathways, as both systems utilize the same G-protein subtype for signal transduction. Mice from both lines were treated with a non-selective CB1/CB2 agonist, WIN55,212-2 and their behavior was evaluated according to the tetrad paradigm assessing antinociception, catalepsy, hypothermia and locomotor activity. Surprisingly, the engagement of CB1 receptors in SSIA was not confirmed. G-protein activation was studied in different brain regions and the spinal cord in the [35S]GTPγS assay. It was shown that WIN55,212-2 produced more potent antinociception in HA than in LA mice. Also, HA mice displayed stronger cannabinoid-induced catalepsy in the bar test. However, LA mice were more sensitive to the hypothermic effect of WIN55,212-2. The intensity of behavioral responses to WIN55,212-2 was correlated with increased G-protein activation in the periaqueductal gray matter, frontal cortex, striatum and thalamus in HA mice. A weak response to WIN55,212-2 in LA mice could depend on impaired CB2 receptor signaling. In conclusion, differences in both opioid and cannabinoid sensitivity between HA and LA mice could stem from alterations in intracellular second messenger mechanisms involving G-protein activation.


Asunto(s)
Encéfalo/fisiología , Dolor/genética , Receptor Cannabinoide CB1/genética , Receptor Cannabinoide CB2/genética , Estrés Psicológico/genética , Analgésicos/farmacología , Analgésicos/uso terapéutico , Animales , Encéfalo/efectos de los fármacos , Proteínas de Unión al GTP/agonistas , Proteínas de Unión al GTP/genética , Proteínas de Unión al GTP/metabolismo , Ratones , Dolor/tratamiento farmacológico , Dolor/metabolismo , Dimensión del Dolor/efectos de los fármacos , Dimensión del Dolor/métodos , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB2/agonistas , Receptor Cannabinoide CB2/metabolismo , Especificidad de la Especie , Estrés Psicológico/metabolismo
14.
Br J Pharmacol ; 176(11): 1649-1663, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30710458

RESUMEN

BACKGROUND AND PURPOSE: The δ-opioid receptor is an emerging target for the management of chronic pain and depression. Biased signalling, the preferential activation of one signalling pathway over another downstream of δ-receptors, may generate better therapeutic profiles. BMS 986187 is a positive allosteric modulator of δ-receptors. Here, we ask if BMS 986187 can directly activate the receptor from an allosteric site, without an orthosteric ligand, and if a signalling bias is generated. EXPERIMENTAL APPROACH: We used several clonal cell lines expressing δ-receptors, to assess effects of BMS 986187 on events downstream of δ-receptors by measuring G-protein activation, ß-arrestin 2 recruitment, receptor phosphorylation, loss of surface receptor expression, ERK1/ERK2 phosphorylation, and receptor desensitization. KEY RESULTS: BMS 986187 is a G protein biased allosteric agonist, relative to ß-arrestin 2 recruitment. Despite showing direct and potent G protein activation, BMS 986187 has a low potency to recruit ß-arrestin 2. This appears to reflect the inability of BMS 986187 to elicit any significant receptor phosphorylation, consistent with low receptor internalization and a slower onset of desensitization, compared with the full agonist SNC80. CONCLUSIONS AND IMPLICATIONS: This is the first evidence of biased agonism mediated through direct binding to an allosteric site on an opioid receptor, without a ligand at the orthosteric site. Our data suggest that agonists targeting δ-receptors, or indeed any GPCR, through allosteric sites may be a novel way to promote signalling bias and thereby potentially produce a more specific pharmacology than can be observed by activation via the orthosteric site.


Asunto(s)
Proteínas de Unión al GTP/agonistas , Receptores Opioides delta/metabolismo , Xantonas/farmacología , Sitio Alostérico , Animales , Células CHO , Cricetulus , Proteínas de Unión al GTP/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Células HEK293 , Humanos , Masculino , Ratones
15.
Basic Clin Pharmacol Toxicol ; 124(6): 649-659, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30507034

RESUMEN

The biochemical abnormalities in transmembrane signal transduction mediated through G protein-coupled receptors (GPCRs) have been postulated as underlying pathophysiology of psychiatric diseases such as schizophrenia and mood disorders. In the present study, the experimental conditions of agonist-induced [35 S]GTPγS binding in postmortem human brain membranes were optimized, and the responses induced by a series of agonists were pharmacologically characterized. The [35 S]GTPγS binding assay was performed in postmortem human prefrontal cortical membranes by means of filtration techniques, and standardized as to GDP concentration, membrane protein content, MgCl2 and NaCl concentrations in assay buffer, incubation period and effect of white matter contamination. Under the standard assay conditions, the specific [35 S]GTPγS binding was stimulated by the addition of 15 compounds in a concentration-dependent manner. Of these agonists, R(+)-8-OH-DPAT, UK-14,304, DAMGO and DPDPE showed apparently biphasic concentration-response curves. As for these four responses, only higher-potency site was pharmacologically characterized. The receptors involved in the responses investigated were 5-HT1A receptor (probed with R(+)-8-OH-DPAT or 5-HT), α2A -adrenoceptor (UK-14,304 or (-)-epinephrine), M2 /M4 mAChRs (carbachol), adenosine A1 receptor (adenosine), histamine H3 receptor (histamine), group II mGlu (l-glutamate), GABAB receptor (baclofen), µ-opioid receptor (DAMGO or endomophin-1), δ-opioid receptor (DPDPE or SNC-80) and NOP (nociceptin). Although dopamine also activated specific [35 S]GTPγS binding, this response was likely mediated via α2A -adrenoceptor, but not dopamine receptor subtypes. The present study provides us with fundamental aspects of the strategy for elucidation of probable abnormalities of neural signalling mediated by G proteins activated through multiple GPCRs in the brain of psychiatric patients.


Asunto(s)
Proteínas de Unión al GTP/agonistas , Proteínas de Unión al GTP/metabolismo , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Corteza Prefrontal/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Unión Competitiva , Femenino , Humanos , Masculino , Persona de Mediana Edad , Unión Proteica , Receptor de Adenosina A1/metabolismo , Receptor de Serotonina 5-HT2C/metabolismo , Receptores Adrenérgicos alfa 2/metabolismo , Receptores de GABA-B/metabolismo , Receptores Histamínicos H3/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Receptores Opioides mu/metabolismo , Receptores sigma/metabolismo , Adulto Joven , Receptor Sigma-1
16.
BMC Immunol ; 19(1): 36, 2018 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-30522434

RESUMEN

BACKGROUND: In coeliac disease, ingestion of gluten induces the production of transglutaminase 2 (TG2)-targeted autoantibodies by TG2-specific plasma cells present at high frequency in the small intestinal mucosa in untreated disease. During treatment with a gluten-free diet (GFD), the number of these cells decreases considerably. It has not been previously investigated whether the cells are also present prior to development of villous atrophy, or in non-responsive patients and those with dietary lapses. We aimed to define the frequency of small bowel mucosal TG2-specific plasma cells in coeliac disease patients with varying disease activity, and to investigate whether the frequency correlates with serum and small intestinal TG2-targeting antibodies as well as mucosal morphology and the number of intraepithelial lymphocytes. RESULTS: Mucosal TG2-specific plasma cells were found in 79% of patients prior to development of mucosal damage, in all patients with villous atrophy, and in 63% of the patients after 1 year on GFD. In these disease stages, TG2-specific plasma cells accounted for median of 2.3, 4.3, and 0.7% of all mucosal plasma cells, respectively. After long-term treatment, the cells were present in 20% of the patients in clinical remission (median 0%) and in 60% of the patients with poor dietary adherence (median 5.8%). In patients with non-responsive coeliac disease despite strict GFD, the cells were found in only one (9%) subject; the cells accounted for 2.4% of all plasma cells. A positive correlation between the percentage of TG2-specific plasma cells and serum TG2 antibody levels (rS = 0.69, P < 0.001) and the intensity of mucosal TG2-targeting IgA deposits (rS = 0.43, P < 0.001) was observed. CONCLUSIONS: Our results show that TG2-specific plasma cells are already detectable prior to villous atrophy, and that generally their frequency increases during overt disease. By contrast, on GFD, the percentage of these cells decreases. Overall, the presence of TG2-specific plasma cells in the small bowel mucosa mirrors the presence of gluten in the diet, but the frequency is not always parallel to the level of serum or intestinal TG2 antibodies. These findings increase the knowledge about the development of the TG2 plasma cell responses especially in the early phases of coeliac disease.


Asunto(s)
Autoanticuerpos/sangre , Enfermedad Celíaca/inmunología , Proteínas de Unión al GTP/agonistas , Mucosa Intestinal/inmunología , Intestino Delgado/inmunología , Células Plasmáticas/inmunología , Adolescente , Adulto , Anciano , Estudios de Cohortes , Dieta Sin Gluten , Femenino , Glútenes/metabolismo , Humanos , Inmunoglobulina A/inmunología , Masculino , Persona de Mediana Edad , Proteína Glutamina Gamma Glutamiltransferasa 2 , Transglutaminasas
19.
J Pharmacol Exp Ther ; 362(2): 254-262, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28533287

RESUMEN

Prescription opioids are a mainstay in the treatment of acute moderate to severe pain. However, chronic use leads to a host of adverse consequences including tolerance and opioid-induced hyperalgesia (OIH), leading to more complex treatment regimens and diminished patient compliance. Patients with OIH paradoxically experience exaggerated nociceptive responses instead of pain reduction after chronic opioid usage. The development of OIH and tolerance tend to occur simultaneously and, thus, present a challenge when studying the molecular mechanisms driving each phenomenon. We tested the hypothesis that a G protein-biased µ-opioid peptide receptor (MOPR) agonist would not induce symptoms of OIH, such as mechanical allodynia, following chronic administration. We observed that the development of opioid-induced mechanical allodynia (OIMA), a model of OIH, was absent in ß-arrestin1-/- and ß-arrestin2-/- mice in response to chronic administration of conventional opioids such as morphine, oxycodone and fentanyl, whereas tolerance developed independent of OIMA. In agreement with the ß-arrestin knockout mouse studies, chronic administration of TRV0109101, a G protein-biased MOPR ligand and structural analog of oliceridine, did not promote the development of OIMA but did result in drug tolerance. Interestingly, following induction of OIMA by morphine or fentanyl, TRV0109101 was able to rapidly reverse allodynia. These observations establish a role for ß-arrestins in the development of OIH, independent of tolerance, and suggest that the use of G protein-biased MOPR ligands, such as oliceridine and TRV0109101, may be an effective therapeutic avenue for managing chronic pain with reduced propensity for opioid-induced hyperalgesia.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Proteínas de Unión al GTP/agonistas , Hiperalgesia/tratamiento farmacológico , Dimensión del Dolor/efectos de los fármacos , Receptores Opioides mu/agonistas , Animales , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Proteínas de Unión al GTP/fisiología , Células HEK293 , Humanos , Hiperalgesia/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Dimensión del Dolor/métodos , Receptores Opioides mu/fisiología
20.
Cell Signal ; 32: 59-65, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28088389

RESUMEN

Nalfurafine is a moderately selective kappa opioid receptor (KOR) analgesic with low incidence of dysphoric side effects in clinical development for the treatment of uremic pruritis. The basis for its reduced dysphoric effect compared to other KOR agonists is not clear, but prior studies suggest that the aversive properties of KOR agonists require p38α MAPK activation through an arrestin-dependent mechanism. To determine whether nalfurafine is a functionally selective KOR agonist, we measured its potency to activate the G protein-dependent early phase of Extracellular Signal-Regulated Kinase (ERK1/2) phosphorylation and the arrestin-dependent late phase of p38 MAPK signaling. Nalfurafine was approximately 250 fold more potent for ERK1/2 activation as compared to p38 MAPK activation in human KOR (hKOR) expressing HEK293 cells, and approximately 20 fold more potent for ERK1/2 activation than p38 activation in rodent KOR (rKOR) expressing HEK293 cells. The 10-fold greater G-bias at the hKOR than rKOR was unexpected, however the G protein biased effect of nalfurafine is consistent with its reduced dysphoric effects in human and rodent models. Although nalfurafine is reported to have low receptor selectivity in radioligand binding assays, its antinociceptive effect was blocked by the selective KOR antagonist norbinaltorphimine. Nalfurafine pretreatment also resulted in a KOR-dependent and mu opioid receptor-independent reduction in scratching induced by 5'-GNTI. These findings suggest that nalfurafine is a functionally selective KOR agonist and that KOR agonists able to selectively activate G protein signaling without activating p38α MAPK may have therapeutic potential as non-dysphoric antipruritic analgesics.


Asunto(s)
Proteínas de Unión al GTP/agonistas , Morfinanos/farmacología , Receptores Opioides kappa/agonistas , Compuestos de Espiro/farmacología , Animales , Tolerancia a Medicamentos , Activación Enzimática/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas de Unión al GTP/metabolismo , Células HEK293 , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Morfinanos/uso terapéutico , Prurito/tratamiento farmacológico , Prurito/patología , Receptores Opioides kappa/metabolismo , Receptores Opioides mu/metabolismo , Transducción de Señal/efectos de los fármacos , Compuestos de Espiro/uso terapéutico , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA