Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Med Chem ; 66(17): 11843-11854, 2023 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-37632447

RESUMEN

The κ-opioid receptor (KOR) is an attractive target for the development of novel drugs. KOR agonists are potentially safer pain medications, whereas KOR antagonists are promising drug candidates for the treatment of neuropsychiatric disorders. Hitherto, the vast majority of selective drug leads that have been developed for KOR are small molecules. In this study, novel peptide probes were designed by using an endogenous dynorphin A1-13 sequence as a template for peptide stapling via late-stage cysteine functionalization. Leveraging this strategy, we developed a stable and potent KOR antagonist, CSD-CH2(1,8)-NH2, with approximately 1000-fold improved selectivity for KOR over µ- and δ-opioid receptors. Its potent competitive KOR antagonism was verified in KOR-expressing cells, peripheral dorsal root ganglion neurons, and using the tail-flick and rotarod tests in mice. This work highlights the value of cysteine stapling to develop selective peptide probes to modulate central KOR function, as innovative peptide drug candidates for the treatment of KOR-related illnesses.


Asunto(s)
Cisteína , Antagonistas de Narcóticos , Animales , Ratones , Péptidos/farmacología , Dinorfinas , Ganglios Espinales , Receptores Opioides kappa
2.
Pharmaceuticals (Basel) ; 14(11)2021 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-34832873

RESUMEN

Opioids are widely used as therapeutic agents against moderate to severe acute and chronic pain. Still, these classes of analgesic drugs have many potential limitations as they induce analgesic tolerance, addiction and numerous behavioural adverse effects that often result in patient non-compliance. As opium and opioids have been traditionally used as painkillers, the exact mechanisms of their adverse reactions over repeated use are multifactorial and not fully understood. Older adults suffer from cancer and non-cancer chronic pain more than younger adults, due to the physiological changes related to ageing and their reduced metabolic capabilities and thus show an increased number of adverse reactions to opioid drugs. All clinically used opioids are µ-opioid receptor agonists, and the major adverse effects are directly or potentially connected to this receptor. Multifunctional opioid ligands or peripherally restricted opioids may elicit fewer adverse effects, as shown in preclinical studies, but these results need reproducibility from further extensive clinical trials. The current review aims to overview various mechanisms involved in the adverse effects induced by opioids, to provide a better understanding of the underlying pathophysiology and, ultimately, to help develop an effective therapeutic strategy to better manage pain.

3.
J Med Chem ; 63(21): 12929-12941, 2020 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-32902268

RESUMEN

Fusion of nonopioid pharmacophores, such as neurotensin, with opioid ligands represents an attractive approach for pain treatment. Herein, the µ-/δ-opioid agonist tetrapeptide H-Dmt-d-Arg-Aba-ß-Ala-NH2 (KGOP01) was fused to NT(8-13) analogues. Since the NTS1 receptor has been linked to adverse effects, selective MOR-NTS2 ligands are preferred. Modifications were introduced within the native NT sequence, particularly a ß3-homo amino acid in position 8 and Tyr11 substitutions. Combination of ß3hArg and Dmt led to peptide 7, a MOR agonist, showing the highest NTS2 affinity described to date (Ki = 3 pM) and good NTS1 affinity (Ki = 4 nM), providing a >1300-fold NTS2 selectivity. The (6-OH)Tic-containing analogue 9 also exhibited high NTS2 affinity (Ki = 1.7 nM), with low NTS1 affinity (Ki = 4.7 µM), resulting in an excellent NTS2 selectivity (>2700). In mice, hybrid 7 produced significant and prolonged antinociception (up to 8 h), as compared to the KGOP01 opioid parent compound.


Asunto(s)
Diseño de Fármacos , Péptidos/química , Receptores de Neurotensina/metabolismo , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Secuencia de Aminoácidos , Animales , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Oligopéptidos/química , Oligopéptidos/metabolismo , Oligopéptidos/uso terapéutico , Dolor/tratamiento farmacológico , Dolor/patología , Péptidos/metabolismo , Péptidos/uso terapéutico , Unión Proteica , Receptores de Neurotensina/química , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Relación Estructura-Actividad
4.
Molecules ; 25(9)2020 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-32365707

RESUMEN

The mu opioid receptor (MOR) is the primary target for analgesia of endogenous opioid peptides, alkaloids, synthetic small molecules with diverse scaffolds, and peptidomimetics. Peptide-based opioids are viewed as potential analgesics with reduced side effects and have received constant scientific interest over the years. This study focuses on three potent peptide and peptidomimetic MOR agonists, DALDA, [Dmt1]DALDA, and KGOP01, and the prototypical peptide MOR agonist DAMGO. We present the first molecular modeling study and structure-activity relationships aided by in vitro assays and molecular docking of the opioid peptide analogues, in order to gain insight into their mode of binding to the MOR. In vitro binding and functional assays revealed the same rank order with KGOP01 > [Dmt1]DALDA > DAMGO > DALDA for both binding and MOR activation. Using molecular docking at the MOR and three-dimensional interaction pattern analysis, we have rationalized the experimental outcomes and highlighted key amino acid residues responsible for agonist binding to the MOR. The Dmt (2',6'-dimethyl-L-Tyr) moiety of [Dmt1]DALDA and KGOP01 was found to represent the driving force for their high potency and agonist activity at the MOR. These findings contribute to a deeper understanding of MOR function and flexible peptide ligand-MOR interactions, that are of significant relevance for the future design of opioid peptide-based analgesics.


Asunto(s)
Oligopéptidos/química , Oligopéptidos/metabolismo , Péptidos/química , Péptidos/metabolismo , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo , Animales , Sitios de Unión , Células CHO , Cricetulus , Humanos , Cinética , Ratones , Modelos Moleculares , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad
5.
Science ; 360(6395)2018 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-29930108

RESUMEN

A systems view of G protein-coupled receptor (GPCR) signaling in its native environment is central to the development of GPCR therapeutics with fewer side effects. Using the kappa opioid receptor (KOR) as a model, we employed high-throughput phosphoproteomics to investigate signaling induced by structurally diverse agonists in five mouse brain regions. Quantification of 50,000 different phosphosites provided a systems view of KOR in vivo signaling, revealing novel mechanisms of drug action. Thus, we discovered enrichment of the mechanistic target of rapamycin (mTOR) pathway by U-50,488H, an agonist causing aversion, which is a typical KOR-mediated side effect. Consequently, mTOR inhibition during KOR activation abolished aversion while preserving beneficial antinociceptive and anticonvulsant effects. Our results establish high-throughput phosphoproteomics as a general strategy to investigate GPCR in vivo signaling, enabling prediction and modulation of behavioral outcomes.


Asunto(s)
Encéfalo/metabolismo , Ensayos Analíticos de Alto Rendimiento , Fosfoproteínas/metabolismo , Proteómica/métodos , Receptores Opioides kappa/metabolismo , Transducción de Señal , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/metabolismo , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Analgésicos no Narcóticos/farmacología , Animales , Anticonvulsivantes/farmacología , Arrestinas/metabolismo , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Línea Celular Tumoral , Diterpenos de Tipo Clerodano/metabolismo , Diterpenos de Tipo Clerodano/farmacología , Humanos , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenetilaminas/metabolismo , Fenetilaminas/farmacología , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/genética , Transducción de Señal/efectos de la radiación , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo
6.
Pain ; 158(3): 505-515, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28135212

RESUMEN

Drugs able to treat both nociceptive and neuropathic pain effectively without major side effects are lacking. We developed a bifunctional peptide-based hybrid (KGNOP1) that structurally combines a mu-opioid receptor agonist (KGOP1) with antinociceptive activity and a weak nociceptin receptor antagonist (KGNOP3) with anti-neuropathic pain activity. We investigated KGNOP1-related behavioral effects after intravenous administration in rats by assessing thermal nociception, cold hyperalgesia in a model of neuropathic pain induced by chronic constriction injury of the sciatic nerve, and plethysmography parameters including inspiratory time (TI) and minute ventilation (VM) in comparison to the well-known opioid analgesics, tramadol and morphine. Time-course and dose-dependent effects were investigated for all behavioral parameters to determine the effective doses 50% (ED50). Pain-related effects on cold hyperalgesia were markedly increased by KGNOP1 as compared to KGNOP3 and tramadol (ED50: 0.0004, 0.32, and 12.1 µmol/kg, respectively), whereas effects on thermal nociception were significantly higher with KGNOP1 as compared to morphine (ED50: 0.41 and 14.7 µmol/kg, respectively). KGNOP1 and KGOP1 produced a larger increase in TI and deleterious decrease in VM in comparison to morphine and tramadol (ED50(TI): 0.63, 0.52, 12.2, and 50.9 µmol/kg; ED50(VM): 0.57, 0.66, 10.6, and 50.0 µmol/kg, respectively). Interestingly, the calculated ratios of anti-neuropathic pain/antinociceptive to respiratory effects revealed that KGNOP1 was safer than tramadol (ED50 ratio: 5.44 × 10 vs 0.24) and morphine (ED50 ratio: 0.72 vs 1.39). We conclude that KGNOP1 is able to treat both experimental neuropathic and nociceptive pain, more efficiently and safely than tramadol and morphine, respectively, and thus should be a candidate for future clinical developments.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Antagonistas de Narcóticos/uso terapéutico , Neuralgia/tratamiento farmacológico , Dolor Nociceptivo/tratamiento farmacológico , Péptidos Opioides/antagonistas & inhibidores , Péptidos/uso terapéutico , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Hiperalgesia/tratamiento farmacológico , Indoles/uso terapéutico , Ligandos , Masculino , Oligopéptidos/química , Oligopéptidos/uso terapéutico , Dimensión del Dolor , Fenalenos/uso terapéutico , Pletismografía , Ratas , Ratas Sprague-Dawley , Receptores Opioides , Respiración/efectos de los fármacos , Factores de Tiempo , Tramadol/uso terapéutico , Nociceptina
7.
Immunol Lett ; 118(1): 82-7, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18440650

RESUMEN

Opioid receptors are expressed not only on neuroendocrine cells but also on immunocompetent cells such as lymphocytes, monocytes and macrophages. micro-Opioid receptor agonists were found to exert immunosuppressive effects, whereas delta-opioid receptor agonists have been shown to act as immunostimulants. delta-Opioid receptor agonists stimulate T and B cells and activate granulocytes and monocytes, conversely, immunostimulation can be blocked by the non-peptidic delta-opioid receptor antagonist (NTI). We investigated the impact of NTI and of the two structurally related compounds HS-378 and HS-459 on degradation of tryptophan and formation of neopterin in mitogen-stimulated human peripheral blood mononuclear cells (PBMC). Both these biochemical pathways were found to be suppressed by all three opioid receptor antagonists, HS-378 and HS-459 exhibiting slightly greater potency than NTI. The suppression of tryptophan degradation suggests that the tested delta-opioid antagonists are able to influence the serotonergic system via a non-opioid action.


Asunto(s)
Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Mitosis/efectos de los fármacos , Receptores Opioides delta/antagonistas & inhibidores , Células Cultivadas , Humanos , Quinurenina/metabolismo , Leucocitos/citología , Estructura Molecular , Péptidos/química , Receptores Opioides delta/metabolismo , Relación Estructura-Actividad , Triptófano/metabolismo
8.
Brain Res Bull ; 74(5): 369-75, 2007 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-17845912

RESUMEN

Peripheral micro-opioid receptors (MOR) have emerged as important components of inhibitory nociceptive pathways. Here, the antinociceptive effects of MOR agonists, the 6beta-glycine derivative of 14-O-methyloxymorphone (HS-731), DAMGO and morphine were evaluated in a mouse model of visceral pain. The abdominal acetic acid-induced writhing test was used to examine the peripheral, preemptive antinociceptive opioid action on visceral nociception. HS-731 administered subcutaneously (s.c.) or intracerebroventricularly (i.c.v.) dose-dependently and completely inhibited writhing, being 24-598-fold more potent, depending on the administration route, than two selective MOR agonists, the enkephalin analogue [D-Ala(2),N-Me-Phe(4),Gly-ol(5)]enkephalin (DAMGO) and morphine. A longer duration of action (2-3 h) was induced by HS-731 given before acetic acid, while shorter effect was produced by morphine (30-60 min) and DAMGO (30-45 min). The antinociceptive effects of systemic opioids were reversed by the s.c. opioid antagonist, naloxone. Blocking of central MOR by the selective MOR antagonist D-Phe-Cys-Tyr-d-Trp-Arg-Thr-Pen-Thr-NH(2) (CTAP, i.c.v.) resulted in a significant reduction of antinociception of s.c. morphine, whereas it completely failed to antagonize the effects of systemic HS-731 or DAMGO. In in vitro studies, HS-731 and DAMGO, but not morphine showed high intrinsic efficacy, naltrexone-sensitive agonist effect at MOR of the rat vas deferens. These data demonstrate that selective activation of peripheral MOR by systemic s.c. HS-731 or DAMGO produces potent peripheral, preemptive visceral antinociception, while morphine's effects are mediated primarily through central mechanisms. Our findings support the role of peripheral MOR in the pathology of pain states involving sensitization of peripheral nociceptors.


Asunto(s)
Analgésicos Opioides/administración & dosificación , Encefalina Ala(2)-MeFe(4)-Gli(5)/administración & dosificación , Compuestos Epoxi/administración & dosificación , Morfinanos/administración & dosificación , Morfina/administración & dosificación , Dolor/tratamiento farmacológico , Conducto Deferente/efectos de los fármacos , Ácido Acético , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Vías de Administración de Medicamentos , Esquema de Medicación , Interacciones Farmacológicas , Técnicas In Vitro , Masculino , Ratones , Dolor/inducido químicamente , Dimensión del Dolor , Fragmentos de Péptidos , Péptidos/uso terapéutico , Ratas , Ratas Wistar , Somatostatina , Factores de Tiempo , Conducto Deferente/fisiología
9.
J Med Chem ; 46(9): 1758-63, 2003 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-12699394

RESUMEN

The synthesis, biological, and pharmacological evaluations of 14beta-O-phenylpropyl-substituted morphinan-6-ones are described. The most striking finding of this study was that all of the compounds from the novel series of differently N-substituted 14beta-O-phenylpropylmorphinans acted as powerful opioid agonists. Even with N-substituents such as cyclopropylmethyl and allyl, which are usually associated with distinct antagonist properties, only agonists were obtained. Compared to morphine, the N-cyclopropylmethyl derivative 15 showed considerably increased potency in the in vivo assays in mice (600-fold in the tail-flick assay, 60-fold in the paraphenylquinone writhing test, and 400-fold in the hot-plate assay). Remarkably, most of the new ligands were nonselective and exhibited binding affinities in the subnanomolar range at opioid receptors (mu, kappa, delta), with the N-propyl derivative 19 displaying the highest affinity for the mu-receptor (K(i) = 0.09 nM).


Asunto(s)
Analgésicos Opioides/síntesis química , Morfinanos/síntesis química , Receptores Opioides/agonistas , Analgésicos Opioides/química , Analgésicos Opioides/farmacología , Animales , Células CHO , Cricetinae , Ligandos , Masculino , Ratones , Ratones Endogámicos ICR , Morfinanos/química , Morfinanos/farmacología , Dimensión del Dolor , Ensayo de Unión Radioligante , Ratas , Receptores Opioides delta/agonistas , Receptores Opioides kappa/agonistas , Receptores Opioides mu/agonistas , Relación Estructura-Actividad , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA