Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 152
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(19)2023 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-37834455

RESUMEN

Pain is the most significant impairment and debilitating challenge for patients with bone metastasis. Therefore, the primary objective of current therapy is to mitigate and prevent the persistence of pain. Thus, cancer-induced bone pain is described as a multifaceted form of discomfort encompassing both inflammatory and neuropathic elements. We have developed a novel non-addictive pain therapeutic, PNA6, that is a derivative of the peptide Angiotensin-(1-7) and binds the Mas receptor to decrease inflammation-related cancer pain. In the present study, we provide evidence that PNA6 attenuates inflammatory, chemotherapy-induced peripheral neuropathy (CIPN) and cancer pain confined to the long bones, exhibiting longer-lasting efficacious therapeutic effects. PNA6, Asp-Arg-Val-Tyr-Ile-His-Ser-(O-ß-Lact)-amide, was successfully synthesized using solid phase peptide synthesis (SPPS). PNA6 significantly reversed inflammatory pain induced by 2% carrageenan in mice. A second murine model of platinum drug-induced painful peripheral neuropathy was established using oxaliplatin. Mice in the oxaliplatin-vehicle treatment groups demonstrated significant mechanical allodynia compared to the oxaliplatin-PNA6 treatment group mice. In a third study modeling a complex pain state, E0771 breast adenocarcinoma cells were implanted into the femur of female C57BL/6J wild-type mice to induce cancer-induced bone pain (CIBP). Both acute and chronic dosing of PNA6 significantly reduced the spontaneous pain behaviors associated with CIBP. These data suggest that PNA6 is a viable lead candidate for treating chronic inflammatory and complex neuropathic pain.


Asunto(s)
Antineoplásicos , Neoplasias Óseas , Neoplasias de la Mama , Dolor en Cáncer , Neuralgia , Humanos , Ratones , Femenino , Animales , Oxaliplatino/efectos adversos , Dolor en Cáncer/tratamiento farmacológico , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Neuralgia/inducido químicamente , Neuralgia/tratamiento farmacológico , Neuralgia/complicaciones , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/complicaciones , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias Óseas/complicaciones , Neoplasias Óseas/tratamiento farmacológico , Antineoplásicos/efectos adversos
2.
Proc Natl Acad Sci U S A ; 116(21): 10557-10562, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31068460

RESUMEN

Neuropathic pain afflicts millions of individuals and represents a major health problem for which there is limited effective and safe therapy. Emerging literature links altered sphingolipid metabolism to nociceptive processing. However, the neuropharmacology of sphingolipid signaling in the central nervous system in the context of chronic pain remains largely unexplored and controversial. We now provide evidence that sphingosine-1-phosphate (S1P) generated in the dorsal horn of the spinal cord in response to nerve injury drives neuropathic pain by selectively activating the S1P receptor subtype 1 (S1PR1) in astrocytes. Accordingly, genetic and pharmacological inhibition of S1PR1 with multiple antagonists in distinct chemical classes, but not agonists, attenuated and even reversed neuropathic pain in rodents of both sexes and in two models of traumatic nerve injury. These S1PR1 antagonists retained their ability to inhibit neuropathic pain during sustained drug administration, and their effects were independent of endogenous opioid circuits. Moreover, mice with astrocyte-specific knockout of S1pr1 did not develop neuropathic pain following nerve injury, thereby identifying astrocytes as the primary cellular substrate of S1PR1 activity. On a molecular level, the beneficial reductions in neuropathic pain resulting from S1PR1 inhibition were driven by interleukin 10 (IL-10), a potent neuroprotective and anti-inflammatory cytokine. Collectively, our results provide fundamental neurobiological insights that identify the cellular and molecular mechanisms engaged by the S1PR1 axis in neuropathic pain and establish S1PR1 as a target for therapeutic intervention with S1PR1 antagonists as a class of nonnarcotic analgesics.


Asunto(s)
Astrocitos/metabolismo , Neuralgia/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Sulfonas/uso terapéutico , Triazoles/uso terapéutico , Animales , Evaluación Preclínica de Medicamentos , Femenino , Interleucina-10/metabolismo , Masculino , Ratones , Neuralgia/tratamiento farmacológico , Neuralgia/etiología , Ratas Sprague-Dawley , Receptores de Esfingosina-1-Fosfato/antagonistas & inhibidores , Sulfonas/farmacología , Triazoles/farmacología
3.
Cephalalgia ; 41(3): 404-416, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33131303

RESUMEN

BACKGROUND: Despite increasing evidence differentiating episodic and chronic migraine, little work has determined how currently utilized animal models of migraine best represent each distinct disease state. AIM: In this review, we seek to characterize accepted preclinical models of migraine-like headache by their ability to recapitulate the clinical allodynic features of either episodic or chronic migraine. METHODS: From a search of the Pu bMed database for "animal models of migraine", "headache models" and "preclinical migraine", we identified approximately 80 recent (within the past 20 years) publications that utilized one of 10 different models for migraine research. Models reviewed fit into one of the following categories: Dural KCl application, direct electrical stimulation, nitroglycerin administration, inflammatory soup injection, CGRP injection, medication overuse, monogenic animals, post-traumatic headache, specific channel activation, and hormone manipulation. Recapitulation of clinical features including cephalic and extracephalic hypersensitivity were evaluated for each and compared. DISCUSSION: Episodic migraineurs comprise over half of the migraine population, yet the vast majority of current animal models of migraine appear to best represent chronic migraine states. While some of these models can be modified to reflect episodic migraine, there remains a need for non-invasive, validated models of episodic migraine to enhance the clinical translation of migraine research.


Asunto(s)
Trastornos Migrañosos , Animales , Modelos Animales de Enfermedad , Cefalea , Hiperalgesia
4.
J Pharmacol Exp Ther ; 374(2): 331-341, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32434943

RESUMEN

Treating chronic pain by using opioids, such as morphine, is hampered by the development of opioid-induced hyperalgesia (OIH; increased pain sensitivity), antinociceptive tolerance, and withdrawal, which can contribute to dependence and abuse. In the central nervous system, the purine nucleoside adenosine has been implicated in beneficial and detrimental actions of morphine, but the extent of their interaction remains poorly understood. Here, we demonstrate that morphine-induced OIH and antinociceptive tolerance in rats is associated with a twofold increase in adenosine kinase (ADK) expression in the dorsal horn of the spinal cord. Blocking ADK activity in the spinal cord provided greater than 90% attenuation of OIH and antinociceptive tolerance through A3 adenosine receptor (A3AR) signaling. Supplementing adenosine signaling with selective A3AR agonists blocked OIH and antinociceptive tolerance in rodents of both sexes. Engagement of A3AR in the spinal cord with an ADK inhibitor or A3AR agonist was associated with reduced dorsal horn of the spinal cord expression of the NOD-like receptor pyrin domain-containing 3 (60%-75%), cleaved caspase 1 (40%-60%), interleukin (IL)-1ß (76%-80%), and tumor necrosis factor (50%-60%). In contrast, the neuroinhibitory and anti-inflammatory cytokine IL-10 increased twofold. In mice, A3AR agonists prevented the development of tolerance in a model of neuropathic pain and reduced naloxone-dependent withdrawal behaviors by greater than 50%. These findings suggest A3AR-dependent adenosine signaling is compromised during sustained morphine to allow the development of morphine-induced adverse effects. These findings raise the intriguing possibility that A3AR agonists may be useful adjunct to opioids to manage their unwanted effects. SIGNIFICANCE STATEMENT: The development of hyperalgesia and antinociceptive tolerance during prolonged opioid use are noteworthy opioid-induced adverse effects that reduce opioid efficacy for treating chronic pain and increase the risk of dependence and abuse. We report that in rodents, these adverse effects are due to reduced adenosine signaling at the A3AR, resulting in NOD-like receptor pyrin domain-containing 3-interleukin-1ß neuroinflammation in spinal cord. These effects are attenuated by A3AR agonists, suggesting that A3AR may be a target for therapeutic intervention with selective A3AR agonist as opioid adjuncts.


Asunto(s)
Analgésicos/efectos adversos , Tolerancia a Medicamentos , Hiperalgesia/inducido químicamente , Morfina/efectos adversos , Receptor de Adenosina A3/metabolismo , Transducción de Señal/efectos de los fármacos , Síndrome de Abstinencia a Sustancias/etiología , Adenosina/metabolismo , Animales , Femenino , Hiperalgesia/metabolismo , Interleucina-10/metabolismo , Interleucina-1beta/biosíntesis , Masculino , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
5.
J Pharmacol Exp Ther ; 369(1): 9-25, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30709867

RESUMEN

Increasing evidence indicates that decreased brain blood flow, increased reactive oxygen species (ROS) production, and proinflammatory mechanisms accelerate neurodegenerative disease progression such as that seen in vascular contributions to cognitive impairment and dementia (VCID) and Alzheimer's disease and related dementias. There is a critical clinical need for safe and effective therapies for the treatment and prevention of cognitive impairment known to occur in patients with VCID and chronic inflammatory diseases such as heart failure (HF), hypertension, and diabetes. This study used our mouse model of VCID/HF to test our novel glycosylated angiotensin-(1-7) peptide Ang-1-6-O-Ser-Glc-NH2 (PNA5) as a therapy to treat VCID and to investigate circulating inflammatory biomarkers that may be involved. We demonstrate that PNA5 has greater brain penetration compared with the native angiotensin-(1-7) peptide. Moreover, after treatment with 1.0/mg/kg, s.c., for 21 days, PNA5 exhibits up to 10 days of sustained cognitive protective effects in our VCID/HF mice that last beyond the peptide half-life. PNA5 reversed object recognition impairment in VCID/HF mice and rescued spatial memory impairment. PNA5 activation of the Mas receptor results in a dose-dependent inhibition of ROS in human endothelial cells. Last, PNA5 treatment decreased VCID/HF-induced activation of brain microglia/macrophages and inhibited circulating tumor necrosis factor α, interleukin (IL)-7, and granulocyte cell-stimulating factor serum levels while increasing that of the anti-inflammatory cytokine IL-10. These results suggest that PNA5 is an excellent candidate and "first-in-class" therapy for treating VCID and other inflammation-related brain diseases.


Asunto(s)
Angiotensina I/química , Angiotensina I/farmacología , Disfunción Cognitiva/complicaciones , Disfunción Cognitiva/tratamiento farmacológico , Demencia Vascular/complicaciones , Memoria/efectos de los fármacos , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacología , Proteínas Proto-Oncogénicas/agonistas , Receptores Acoplados a Proteínas G/agonistas , Angiotensina I/farmacocinética , Angiotensina I/uso terapéutico , Animales , Conducta Animal/efectos de los fármacos , Biomarcadores/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/fisiopatología , Electrocardiografía , Glicosilación , Semivida , Insuficiencia Cardíaca/complicaciones , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Inflamación/fisiopatología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Fragmentos de Péptidos/farmacocinética , Fragmentos de Péptidos/uso terapéutico , Proto-Oncogenes Mas , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Memoria Espacial/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos
6.
J Neurosci ; 35(15): 6057-67, 2015 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-25878279

RESUMEN

More than 1.5 billion people worldwide suffer from chronic pain, yet current treatment strategies often lack efficacy or have deleterious side effects in patients. Adenosine is an inhibitory neuromodulator that was previously thought to mediate antinociception through the A1 and A2A receptor subtypes. We have since demonstrated that A3AR agonists have potent analgesic actions in preclinical rodent models of neuropathic pain and that A3AR analgesia is independent of adenosine A1 or A2A unwanted effects. Herein, we explored the contribution of the GABA inhibitory system to A3AR-mediated analgesia using well-characterized mouse and rat models of chronic constriction injury (CCI)-induced neuropathic pain. The deregulation of GABA signaling in pathophysiological pain states is well established: GABA signaling can be hampered by a reduction in extracellular GABA synthesis by GAD65 and enhanced extracellular GABA reuptake via the GABA transporter, GAT-1. In neuropathic pain, GABAAR-mediated signaling can be further disrupted by the loss of the KCC2 chloride anion gradient. Here, we demonstrate that A3AR agonists (IB-MECA and MRS5698) reverse neuropathic pain via a spinal mechanism of action that modulates GABA activity. Spinal administration of the GABAA antagonist, bicuculline, disrupted A3AR-mediated analgesia. Furthermore, A3AR-mediated analgesia was associated with reductions in CCI-related GAD65 and GAT-1 serine dephosphorylation as well as an enhancement of KCC2 serine phosphorylation and activity. Our results suggest that A3AR-mediated reversal of neuropathic pain increases modulation of GABA inhibitory neurotransmission both directly and indirectly through protection of KCC2 function, underscoring the unique utility of A3AR agonists in chronic pain.


Asunto(s)
Agonistas del Receptor de Adenosina A3/uso terapéutico , Analgésicos/uso terapéutico , Ciática/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Simportadores/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacología , Adenosina/uso terapéutico , Animales , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/etiología , Masculino , Ratones , Umbral del Dolor/efectos de los fármacos , Piridinas/farmacología , Piridinas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Receptores de GABA-A/genética , Receptores de GABA-A/metabolismo , Ciática/complicaciones , Transducción de Señal/fisiología , Raíces Nerviosas Espinales/metabolismo , Raíces Nerviosas Espinales/patología , Tiazoles/farmacología , Tiazoles/uso terapéutico , Tioglicolatos/farmacología , Tioglicolatos/uso terapéutico , Cotransportadores de K Cl
7.
Bioorg Med Chem Lett ; 26(1): 222-7, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26611918

RESUMEN

N-Phenyl-N-(piperidin-2-ylmethyl)propionamide based bivalent ligands are unexplored for the design of opioid based ligands. Two series of hybrid molecules bearing N-phenyl-N-(piperidin-2-ylmethyl)propionamide derived small molecules conjugated with an enkephalin analogues with and without a linker (ß-alanine) were designed and synthesized. Both bivalent ligand series exhibited remarkable binding affinities from nanomolar to subnanomolar range at both µ and δ opioid receptors and displayed potent agonist activities as well. The replacement of Tyr with Dmt and introduction of a linker between the small molecule and enkephalin analogue resulted in highly potent ligands. Both series of ligands showed excellent binding affinities at both µ (0.6-0.9nM) and δ (0.2-1.2nM) opioid receptors respectively. Similarly, these bivalent ligands exhibited potent agonist activities in both MVD and GPI assays. Ligand 17 was evaluated for in vivo antinociceptive activity in non-injured rats following spinal administration. Ligand 17 was not significantly effective in alleviating acute pain. The most likely explanations for this low intrinsic efficacy in vivo despite high in vitro binding affinity, moderate in vitro activity are (i) low potency suggesting that higher doses are needed; (ii) differences in experimental design (i.e. non-neuronal, high receptor density for in vitro preparations versus CNS site of action in vitro); (iii) pharmacodynamics (i.e. engaging signalling pathways); (iv) pharmacokinetics (i.e. metabolic stability). In summary, our data suggest that further optimisation of this compound 17 is required to enhance intrinsic antinociceptive efficacy.


Asunto(s)
Amidas/síntesis química , Amidas/farmacología , Analgésicos/síntesis química , Analgésicos/farmacología , Encefalinas/química , Encefalinas/farmacología , Dolor/tratamiento farmacológico , Piperidinas/síntesis química , Piperidinas/farmacología , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Amidas/química , Analgésicos/química , Animales , Relación Dosis-Respuesta a Droga , Encefalinas/síntesis química , Cobayas , Humanos , Íleon/efectos de los fármacos , Ligandos , Ratones , Estructura Molecular , Piperidinas/química , Ratas , Relación Estructura-Actividad
8.
Bioorg Med Chem ; 24(2): 85-91, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26712115

RESUMEN

Here, we report the design, synthesis and structure activity relationship of novel small molecule opioid ligands based on 5-amino substituted (tetrahydronaphthalen-2-yl)methyl moiety with N-phenyl-N-(piperidin-2-yl)propionamide derivatives. We synthesized various molecules including amino, amide and hydroxy substitution on the 5th position of the (tetrahydronaphthalen-2-yl)methyl moiety. In our further designs we replaced the (tetrahydronaphthalen-2-yl)methyl moiety with benzyl and phenethyl moiety. These N-phenyl-N-(piperidin-2-yl)propionamide analogues showed moderate to good binding affinities (850-4 nM) and were selective towards the µ opioid receptor over the δ opioid receptors. From the structure activity relationship studies, we found that a hydroxyl substitution at the 5th position of (tetrahydronapthalen-2yl)methyl group, ligands 19 and 20, showed excellent binding affinities 4 and 5 nM, respectively, and 1000 fold selectivity towards the µ opioid relative to the delta opioid receptor. The ligand 19 showed potent agonist activities 75±21 nM, and 190±42 nM in the GPI and MVD assays. Surprisingly the fluoro analogue 20 showed good agonist activities in MVD assays 170±42 nM, in contrast to its binding affinity results.


Asunto(s)
Amidas/química , Amidas/farmacología , Analgésicos Opioides/química , Analgésicos Opioides/farmacología , Diseño de Fármacos , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo , Amidas/síntesis química , Analgésicos Opioides/síntesis química , Relación Dosis-Respuesta a Droga , Humanos , Ligandos , Estructura Molecular , Relación Estructura-Actividad
9.
Brain ; 138(Pt 1): 28-35, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25414036

RESUMEN

Chronic pain is a global burden that promotes disability and unnecessary suffering. To date, efficacious treatment of chronic pain has not been achieved. Thus, new therapeutic targets are needed. Here, we demonstrate that increasing endogenous adenosine levels through selective adenosine kinase inhibition produces powerful analgesic effects in rodent models of experimental neuropathic pain through the A3 adenosine receptor (A3AR, now known as ADORA3) signalling pathway. Similar results were obtained by the administration of a novel and highly selective A3AR agonist. These effects were prevented by blockade of spinal and supraspinal A3AR, lost in A3AR knock-out mice, and independent of opioid and endocannabinoid mechanisms. A3AR activation also relieved non-evoked spontaneous pain behaviours without promoting analgesic tolerance or inherent reward. Further examination revealed that A3AR activation reduced spinal cord pain processing by decreasing the excitability of spinal wide dynamic range neurons and producing supraspinal inhibition of spinal nociception through activation of serotonergic and noradrenergic bulbospinal circuits. Critically, engaging the A3AR mechanism did not alter nociceptive thresholds in non-neuropathy animals and therefore produced selective alleviation of persistent neuropathic pain states. These studies reveal A3AR activation by adenosine as an endogenous anti-nociceptive pathway and support the development of A3AR agonists as novel therapeutics to treat chronic pain.


Asunto(s)
Neuralgia/metabolismo , Neuronas/metabolismo , Receptor de Adenosina A3/metabolismo , Médula Espinal/metabolismo , Adenosina/farmacología , Animales , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Hiperalgesia/diagnóstico , Hiperalgesia/fisiopatología , Masculino , Bulbo Raquídeo/efectos de los fármacos , Bulbo Raquídeo/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Morfolinas/farmacología , Morfolinas/uso terapéutico , Naloxona/administración & dosificación , Neuralgia/tratamiento farmacológico , Neuralgia/genética , Neuralgia/patología , Neuronas/efectos de los fármacos , Dimensión del Dolor , Umbral del Dolor/efectos de los fármacos , Antagonistas de Receptores Purinérgicos P1/farmacología , Piridinas/farmacología , Piridinas/uso terapéutico , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Receptor de Adenosina A3/genética , Médula Espinal/efectos de los fármacos , Médula Espinal/patología , Factores de Tiempo
10.
J Pharmacol Exp Ther ; 352(3): 462-70, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25512370

RESUMEN

Morphine is metabolized in humans to morphine-3-glucuronide (M3G) and the pharmacologically active morphine-6-glucuronide (M6G). The hepatobiliary disposition of both metabolites relies upon multidrug resistance-associated proteins Mrp3 and Mrp2, located on the sinusoidal and canalicular membrane, respectively. Nonalcoholic steatohepatitis (NASH), the severe stage of nonalcoholic fatty liver disease, alters xenobiotic metabolizing enzyme and transporter function. The purpose of this study was to determine whether NASH contributes to the large interindividual variability and postoperative adverse events associated with morphine therapy. Male Sprague-Dawley rats were fed a control diet or a methionine- and choline-deficient diet to induce NASH. Radiolabeled morphine (2.5 mg/kg, 30 µCi/kg) was administered intravenously, and plasma and bile (0-150 or 0-240 minutes), liver and kidney, and cumulative urine were analyzed for morphine and M3G. The antinociceptive response to M6G (5 mg/kg) was assessed (0-12 hours) after direct intraperitoneal administration since rats do not produce M6G. NASH caused a net decrease in morphine concentrations in the bile and plasma and a net increase in the M3G/morphine plasma area under the concentration-time curve ratio, consistent with upregulation of UDP-glucuronosyltransferase Ugt2b1. Despite increased systemic exposure to M3G, NASH resulted in decreased biliary excretion and hepatic accumulation of M3G. This shift toward systemic retention is consistent with the mislocalization of canalicular Mrp2 and increased expression of sinusoidal Mrp3 in NASH and may correlate to increased antinociception by M6G. Increased metabolism and altered transporter regulation in NASH provide a mechanistic basis for interindividual variability in morphine disposition that may lead to opioid-related toxicity.


Asunto(s)
Derivados de la Morfina/sangre , Morfina/administración & dosificación , Morfina/sangre , Enfermedad del Hígado Graso no Alcohólico/sangre , Animales , Infusiones Intravenosas , Masculino , Enfermedad del Hígado Graso no Alcohólico/patología , Ratas , Ratas Sprague-Dawley , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología
11.
Bioorg Med Chem Lett ; 25(20): 4683-8, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26323872

RESUMEN

We describe the design and synthesis of novel bivalent ligands based on the conjugation of 4-anilidopiperidine derivatives with enkephalin analogues. The design of non-peptide analogues is explored with 5-amino substituted (tetrahydronaphthalen-2yl) methyl containing 4-anilidopiperidine derivatives, while non-peptide-peptide ligands are explored by conjugating the C-terminus of enkephalin analogues (H-Xxx-DAla-Gly-Phe-OH) to the amino group of 4-anilidopiperidine small molecule derivatives with and without a linker. These novel bivalent ligands are evaluated for biological activities at µ and δ opioid receptors. They exhibit very good affinities at µ and δ opioid receptors, and potent agonist activities in MVD and GPI assays. Among these the lead bivalent ligand 17 showed excellent binding affinities (0.1 nM and 0.5 nM) at µ and δ opioid receptors respectively, and was found to have very potent agonist activities in MVD (56 ± 5.9 nM) and GPI (4.6 ± 1.9 nM) assays. In vivo the lead bivalent ligand 17 exhibited a short duration of action (<15 min) comparable to 4-anilidopiperidine derivatives, and moderate analgesic activity. The ligand 17 has limited application against acute pain but may have utility in settings where a highly reversible analgesic is required.


Asunto(s)
Analgésicos/farmacología , Diseño de Fármacos , Encefalinas/farmacología , Contracción Muscular/efectos de los fármacos , Umbral del Dolor/efectos de los fármacos , Piperidinas/farmacología , Receptores Opioides/agonistas , Analgésicos/administración & dosificación , Analgésicos/química , Animales , Relación Dosis-Respuesta a Droga , Encefalinas/administración & dosificación , Encefalinas/química , Cobayas , Ligandos , Ratones , Conformación Molecular , Dimensión del Dolor/efectos de los fármacos , Piperidinas/administración & dosificación , Piperidinas/química , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad
12.
Bioorg Med Chem Lett ; 25(17): 3716-20, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26212775

RESUMEN

Several bifunctional peptides were synthesized and characterized based on the pentapeptide-derived ligand NP30 (1: Tyr-DAla-Gly-Phe-Gly-Trp-O-[3',5'-Bzl(CF3)2]). Modification and truncation of amino acid residues were performed, and the tripeptide-derived ligand NP66 (11: Dmt-DAla-Trp-NH-[3',5'-(CF3)2-Bzl]) was obtained based on the overlapping pharmacophore concept. The Trp(3) residue of ligand 11 works as a message residue for both opioid and NK1 activities. The significance lies in the observation that the approach of appropriate truncation of peptide sequence could lead to a tripeptide-derived chimeric ligand with effective binding and functional activities for both mu and delta opioid and NK1 receptors with agonist activities at mu and delta opioid and antagonist activity at NK1 receptors, respectively.


Asunto(s)
Antagonistas del Receptor de Neuroquinina-1/farmacología , Péptidos/química , Péptidos/farmacología , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Animales , Técnicas de Química Sintética , Descubrimiento de Drogas , Evaluación Preclínica de Medicamentos/métodos , Guanosina 5'-O-(3-Tiotrifosfato) , Humanos , Concentración 50 Inhibidora , Ligandos , Antagonistas del Receptor de Neuroquinina-1/química , Péptidos/metabolismo , Ratas , Relación Estructura-Actividad , Triptófano/química , Triptófano/metabolismo
13.
Bioorg Med Chem ; 23(18): 6185-94, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26299827

RESUMEN

A new series of novel opioid ligands have been designed and synthesized based on the 4-anilidopiperidine scaffold containing a 5-substituted tetrahydronaphthalen-2yl)methyl group with different N-phenyl-N-(piperidin-4-yl)propionamide derivatives to study the biological effects of these substituents on µ and δ opioid receptor interactions. Recently our group reported novel 4-anilidopiperidine analogues, in which several aromatic ring-contained amino acids were conjugated with N-phenyl-N-(piperidin-4-yl)propionamide and examined their biological activities at the µ and δ opioid receptors. In continuation of our efforts in these novel 4-anilidopiperidine analogues, we took a peptidomimetic approach in the present design, in which we substituted aromatic amino acids with tetrahydronaphthalen-2yl methyl moiety with amino, amide and hydroxyl substitutions at the 5th position. In in vitro assays these ligands, showed very good binding affinity and highly selective toward the µ opioid receptor. Among these, the lead ligand 20 showed excellent binding affinity (2 nM) and 5000 fold selectivity toward the µ opioid receptor, as well as functional selectivity in GPI assays (55.20 ± 4.30 nM) and weak or no agonist activities in MVD assays. Based on the in vitro bioassay results the lead compound 20 was chosen for in vivo assessment for efficacy in naïve rats after intrathecal administration. Compound 20 was not significantly effective in alleviating acute pain. This discrepancy between high in vitro binding affinity, moderate in vitro activity, and low in vivo activity may reflect differences in pharmacodynamics (i.e., engaging signaling pathways) or pharmacokinetics (i.e., metabolic stability). In sum, our data suggest that further optimization of this compound 20 is required to enhance in vivo activity.


Asunto(s)
Amidas/química , Receptores Opioides/química , Amidas/síntesis química , Amidas/farmacocinética , Analgésicos Opioides/síntesis química , Analgésicos Opioides/química , Analgésicos Opioides/farmacología , Animales , Conducta Animal/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Semivida , Ligandos , Masculino , Antagonistas de Narcóticos/síntesis química , Antagonistas de Narcóticos/química , Antagonistas de Narcóticos/farmacocinética , Unión Proteica , Ratas , Ratas Sprague-Dawley , Receptores Opioides/metabolismo , Receptores Opioides delta/química , Receptores Opioides delta/metabolismo , Receptores Opioides mu/química , Receptores Opioides mu/metabolismo
14.
J Am Chem Soc ; 136(18): 6608-16, 2014 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-24742335

RESUMEN

We hypothesized that under chronic pain conditions, up-regulated dynorphin A (Dyn A) interacts with bradykinin receptors (BRs) in the spinal cord to promote hyperalgesia through an excitatory effect, which is opposite to the well-known inhibitory effect of opioid receptors. Considering the structural dissimilarity between Dyn A and endogenous BR ligands, bradykinin (BK) and kallidin (KD), this interaction could not be predicted, but it allowed us to discover a potential neuroexcitatory target. Well-known BR ligands, BK, [des-Arg(10), Leu(9)]-kallidin (DALKD), and HOE140 showed different binding profiles at rat brain BRs than that previously reported. These results suggest that neuronal BRs in the rat central nervous system (CNS) may be pharmacologically distinct from those previously defined in non-neuronal tissues. Systematic structure-activity relationship (SAR) study at the rat brain BRs was performed, and as a result, a new key structural feature of Dyn A for BR recognition was identified: amphipathicity. NMR studies of two lead ligands, Dyn A-(4-11) 7 and [des-Arg(7)]-Dyn A-(4-11) 14, which showed the same high binding affinity, confirmed that the Arg residue in position 7, which is known to be crucial for Dyn A's biological activity, is not necessary, and that a type I ß-turn structure at the C-terminal part of both ligands plays an important role in retaining good binding affinities at the BRs. Our lead ligand 14 blocked Dyn A-(2-13) 10-induced hyperalgesic effects and motor impairment in in vivo assays using naïve rats. In a model of peripheral neuropathy, intrathecal (i.th.) administration of ligand 14 reversed thermal hyperalgesia and mechanical hypersensitivity in a dose-dependent manner in nerve-injured rats. Thus, ligand 14 may inhibit abnormal pain states by blocking the neuroexcitatory effects of enhanced levels of Dyn A, which are likely to be mediated by BRs in the spinal cord.


Asunto(s)
Dinorfinas/farmacología , Receptores de Bradiquinina/metabolismo , Médula Espinal/efectos de los fármacos , Animales , Dinorfinas/química , Espectroscopía de Resonancia Magnética , Masculino , Ensayo de Unión Radioligante , Ratas , Ratas Sprague-Dawley , Médula Espinal/metabolismo , Relación Estructura-Actividad
15.
Artículo en Inglés | MEDLINE | ID: mdl-38929040

RESUMEN

Despite the lack of evidence, opioids are still routinely used as a solution to long-term management for chronic noncancer pain (CNCP). Given the significant risks associated with long-term opioid use, including the increased number of unregulated opioid pills at large in the opioid ecosystem, opioid cessation or reduction may be the desired goal of the patient and clinician. Viable nonpharmacological interventions (NPIs) to complement and/or replace opioids for CNCP are needed. Comprehensive reviews that address the impact of NPIs to help adults with CNCP reduce opioid use safely are lacking. We conducted a literature search in PubMed, CINAHL, Embase, PsycINFO, and Scopus for studies published in English. The initial search was conducted in April 2021, and updated in January 2024. The literature search yielded 19,190 relevant articles. Thirty-nine studies met the eligibility criteria and underwent data extraction. Of these, nineteen (49%) were randomized controlled trials, eighteen (46%) were observational studies, and two (5%) were secondary analyses. Among adults with CNCP who use opioids for pain management, studies on mindfulness, yoga, educational programs, certain devices or digital technology, chiropractic, and combination NPIs suggest that they might be an effective approach for reducing both pain intensity and opioid use, but other NPIs did not show a significant effect (e.g., hypnosis, virtual reality). This review revealed there is a small to moderate body of literature demonstrating that some NPIs might be an effective and safe approach for reducing pain and opioid use, concurrently.


Asunto(s)
Analgésicos Opioides , Dolor Crónico , Humanos , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/terapia , Analgésicos Opioides/uso terapéutico , Trastornos Relacionados con Opioides , Manejo del Dolor/métodos
16.
Pain ; 165(3): 666-673, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-37733475

RESUMEN

ABSTRACT: Co-occurrence of chronic pain and clinically significant symptoms of anxiety and/or depression is regularly noted in the literature. Yet, little is known empirically about population prevalence of co-occurring symptoms, nor whether people with co-occurring symptoms constitute a distinct subpopulation within US adults living with chronic pain or US adults living with anxiety and/or depression symptoms (A/D). To address this gap, this study analyzes data from the 2019 National Health Interview Survey, a representative annual survey of self-reported health status and treatment use in the United States (n = 31,997). Approximately 12 million US adults, or 4.9% of the adult population, have co-occurring chronic pain and A/D symptoms. Unremitted A/D symptoms co-occurred in 23.9% of US adults with chronic pain, compared with an A/D prevalence of 4.9% among those without chronic pain. Conversely, chronic pain co-occurred in the majority (55.6%) of US adults with unremitted A/D symptoms, compared with a chronic pain prevalence of 17.1% among those without A/D symptoms. The likelihood of experiencing functional limitations in daily life was highest among those experiencing co-occurring symptoms, compared with those experiencing chronic pain alone or A/D symptoms alone. Among those with co-occurring symptoms, 69.4% reported that work was limited due to a health problem, 43.7% reported difficulty doing errands alone, and 55.7% reported difficulty participating in social activities. These data point to the need for targeted investment in improving functional outcomes for the nearly 1 in 20 US adults living with co-occurring chronic pain and clinically significant A/D symptoms.


Asunto(s)
Dolor Crónico , Depresión , Adulto , Humanos , Estados Unidos/epidemiología , Depresión/epidemiología , Dolor Crónico/epidemiología , Prevalencia , Ansiedad/epidemiología , Trastornos de Ansiedad
17.
Mol Pharmacol ; 84(5): 774-86, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24019224

RESUMEN

Effective pharmacologic treatment of pain with opioids requires that these drugs attain efficacious concentrations in the central nervous system (CNS). A primary determinant of CNS drug permeation is P-glycoprotein (P-gp), an endogenous blood-brain barrier (BBB) efflux transporter that is involved in brain-to-blood transport of opioid analgesics (i.e., morphine). Recently, the nuclear receptor constitutive androstane receptor (CAR) has been identified as a regulator of P-gp functional expression at the BBB. This is critical to pharmacotherapy of pain/inflammation, as patients are often administered acetaminophen (APAP), a CAR-activating ligand, in conjunction with an opioid. Our objective was to investigate, in vivo, the role of CAR in regulation of P-gp at the BBB. Following APAP treatment, P-gp protein expression was increased up to 1.4-1.6-fold in a concentration-dependent manner. Additionally, APAP increased P-gp transport of BODIPY-verapamil in freshly isolated rat brain capillaries. This APAP-induced increase in P-gp expression and activity was attenuated in the presence of CAR pathway inhibitor okadaic acid or transcriptional inhibitor actinomycin D, suggesting P-gp regulation is CAR-dependent. Furthermore, morphine brain accumulation was enhanced by P-gp inhibitors in APAP-treated animals, suggesting P-gp-mediated transport. A warm-water (50°C) tail-flick assay revealed a significant decrease in morphine analgesia in animals treated with morphine 3 or 6 hours after APAP treatment, as compared with animals treated concurrently. Taken together, our data imply that inclusion of APAP in a pain treatment regimen activates CAR at the BBB and increases P-gp functional expression, a clinically significant drug-drug interaction that modulates opioid analgesic efficacy.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Acetaminofén/farmacología , Analgésicos no Narcóticos/farmacología , Barrera Hematoencefálica/metabolismo , Receptores Citoplasmáticos y Nucleares/fisiología , Animales , Transporte Biológico/efectos de los fármacos , Encéfalo/irrigación sanguínea , Receptor de Androstano Constitutivo , Femenino , Morfina/farmacocinética , Permeabilidad , Ratas , Ratas Sprague-Dawley
18.
Bioorg Med Chem Lett ; 23(11): 3434-7, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23623418

RESUMEN

We report here the design, synthesis, and in vitro characterization of new opioid peptides featuring a 4-anilidopiperidine moiety. Despite the fact that the chemical structures of fentanyl surrogates have been found suboptimal per se for the opioid activity, the corresponding conjugates with opioid peptides displayed potent opioid activity. These studies shed an instructive light on the strategies and potential therapeutic values of anchoring the 4-anilidopiperidine scaffold to different classes of opioid peptides.


Asunto(s)
Péptidos Opioides/química , Piperidinas/química , Animales , Línea Celular , Fentanilo/química , Humanos , Cinética , Ratones , Péptidos Opioides/síntesis química , Péptidos Opioides/metabolismo , Piperidinas/síntesis química , Piperidinas/metabolismo , Unión Proteica , Receptores Opioides delta/química , Receptores Opioides delta/genética , Receptores Opioides delta/metabolismo , Receptores Opioides mu/química , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Relación Estructura-Actividad
19.
Bioorg Med Chem Lett ; 23(17): 4975-8, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23899615

RESUMEN

The optimization and truncation of our lead peptide-derived ligand TY005 possessing eight amino-acid residues was performed. Among the synthesized derivatives, NP30 (Tyr(1)-DAla(2)-Gly(3)-Phe(4)-Gly(5)-Trp(6)-O-[3',5'-Bzl(CF3)2]) showed balanced and potent opioid agonist as well as substance P antagonist activities in isolated tissue-based assays, together with significant antinociceptive and antiallodynic activities in vivo.


Asunto(s)
Antagonistas del Receptor de Neuroquinina-1/química , Antagonistas del Receptor de Neuroquinina-1/farmacología , Péptidos/química , Péptidos/farmacología , Receptores Opioides delta/agonistas , Receptores Opioides mu/agonistas , Secuencia de Aminoácidos , Analgésicos/química , Analgésicos/farmacología , Analgésicos/uso terapéutico , Animales , Humanos , Ligandos , Antagonistas del Receptor de Neuroquinina-1/uso terapéutico , Péptidos/uso terapéutico , Ratas , Receptores de Neuroquinina-1/metabolismo , Receptores Opioides delta/metabolismo , Receptores Opioides mu/metabolismo
20.
Med Educ Online ; 28(1): 2167258, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36642963

RESUMEN

Pre-clerkship curricula of most Liaison Committee on Medical Education (LCME)-accredited medical schools are divided into blocks by organ system, leaving a significant amount of information susceptible to loss due to prolonged nonuse. We describe the implementation of a formal Spiral Curriculum that periodically revisits material from previous blocks. Learners were surveyed on receptivity to the curriculum across three graduating classes at a single medical school. Medical school graduate classes of 2020, 2021, and 2022 were surveyed at the end of their pre-clerkship years (2018-2020). The class of 2022 actually received the Spiraled Curriculum intervention, for which the authors created 500 board exam style multiple-choice questions, periodically administered via mandatory in-class sessions ranging from 10 to 20 questions reviewing content from previous blocks with designated expert faculty. Response rates were 36% (n = 46), 45% (n = 52), and 32% (n = 40) for classes of 2020, 2021, and 2022, respectively. On a Likert scale (1 = strongly disagree, 5 = neutral, 10 = strongly agree), the classes of 2020, 2021, and 2022 provided statistically significant differences in their belief that a Spiraled Curriculum would/did help them retain information as 8.2 (SD 1.7), 8.2 (SD 2.2), and 5.0 (SD 3.0) (n < 0.05). All classes endorsed neutral confidence in the existing pre-clerkship curriculum in themselves to prepare for United Stated Medical Licensing Examination (USMLE) Step 1, and in their retention of previous block material with no statistically significant differences between classes. USMLE Step 1 scores did not differ significantly between classes (n = 0.21). Those who did not receive the Spiral Curriculum were highly receptive to it in theory, while those who actually received the intervention gave a neutral rating. Per survey comments, implementation of a Spiraling Curriculum would ideally be administered as either team-based or self-directed activities, and a Spiraling Curriculum may be difficult to implement in accelerated (18 month) pre-clerkship formats.Practice points Question: What is the receptivity of medical students to a formal Spiral curriculum that uses time-spaced repetition sessions of board exam style questions to revisit previous block content of their pre-clerkship years?Findings: In this single-center, quasi-experimental study, the two control group medical school classes had very positive theoretical reception to a Spiral curriculum proposal (rated 8 out of 10) while the class who actually received the Spiral curriculum provided a statistically significant lower neutral rating (rated 5 out of 10), citing preference for a team-based or self-directed format.Meaning: Medical students are strongly in favor of structured time-spaced repetition with board exam style questions to revisit previous material but prefer a format that does not interfere with time to personalize their medical school experience.


Asunto(s)
Prácticas Clínicas , Educación de Pregrado en Medicina , Educación Médica , Estudiantes de Medicina , Humanos , Facultades de Medicina , Curriculum , Evaluación Educacional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA