Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
1.
Int J Mol Sci ; 24(3)2023 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-36769126

RESUMEN

Studies performed in a mouse model of chronic inflammatory pain induced by intraplantar injection of complete Freund's adjuvant (CFA) have shown that constitutive activation of the endogenous opioid signaling, besides serving as a mechanism of endogenous analgesia that tonically represses pain sensitization, also generates a state of endogenous opioid dependence. Since species-related differences concerning pain biology and addictive behaviors occur between mice and rats, the present study explored whether the coexistence of endogenous opioid analgesia and endogenous opioid dependence also characterizes a homologous rat model. To this aim, CFA-injured Wistar rats were treated with either 3 mg/kg or 10 mg/kg of the opioid receptor inverse agonist naltrexone (NTX) during the pain remission phase and monitored for 60 min for possible withdrawal behaviors. At 3 mg/kg, NTX, besides inducing the reinstatement of mechanical allodynia, also caused a distinct appearance of ptosis, with slight but nonsignificant changes to the occurrence of teeth chatters and rearing. On the other hand, 10 mg/kg of NTX failed to unmask pain sensitization and induced significantly lower levels of ptosis than 3 mg/kg. Such an NTX-related response pattern observed in the rat CFA model seems to differ substantially from the pattern previously described in the mouse CFA model. This supports the knowledge that mice and rats are not identical in terms of pharmacological response and stresses the importance of choosing the appropriate species for preclinical pain research purposes depending on the scientific question being asked.


Asunto(s)
Dolor Crónico , Trastornos Relacionados con Opioides , Ratas , Ratones , Animales , Analgésicos Opioides/farmacología , Agonismo Inverso de Drogas , Ratas Wistar , Inflamación/tratamiento farmacológico , Dolor Crónico/tratamiento farmacológico , Hiperalgesia/tratamiento farmacológico , Péptidos Opioides/uso terapéutico , Naltrexona/farmacología , Naltrexona/uso terapéutico , Trastornos Relacionados con Opioides/tratamiento farmacológico , Modelos Animales de Enfermedad
2.
Int J Mol Sci ; 22(24)2021 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-34948415

RESUMEN

Opioids are the most potent widely used analgesics, primarily, but not exclusively, in palliative care. However, they are associated with numerous side effects, such as tolerance, addiction, respiratory depression, and cardiovascular events. This, in turn, can result in their overuse in cases of addiction, the need for dose escalation in cases of developing tolerance, and the emergence of dose-related opioid toxicity, resulting in respiratory depression or cardiovascular problems that can even lead to unintentional death. Therefore, a very important challenge for researchers is to look for ways to counteract the side effects of opioids. The use of peptides and their related compounds, which have been shown to modulate the effects of opioids, may provide such an opportunity. This short review is a compendium of knowledge about the most important and recent findings regarding selected peptides and their modulatory effects on various opioid actions, including cardiovascular and respiratory responses. In addition to the peptides more commonly reported in the literature in the context of their pro- and/or anti-opioid activity-such as neuropeptide FF (NPFF), cholecystokinin (CCK), and melanocyte inhibiting factor (MIF)-we also included in the review nociceptin/orphanin (N/OFQ), ghrelin, oxytocin, endothelin, and venom peptides.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Antagonistas de Narcóticos/uso terapéutico , Péptidos/uso terapéutico , Analgésicos Opioides/farmacología , Animales , Colecistoquinina/farmacología , Colecistoquinina/uso terapéutico , Tolerancia a Medicamentos , Ghrelina/farmacología , Ghrelina/uso terapéutico , Humanos , Antagonistas de Narcóticos/farmacología , Oligopéptidos/farmacología , Oligopéptidos/uso terapéutico , Péptidos Opioides/farmacología , Péptidos Opioides/uso terapéutico , Péptidos/farmacología , Receptores Opioides/metabolismo , Nociceptina
3.
Pharmacol Res ; 158: 104855, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32438036

RESUMEN

Hemorphins are endogenous peptides, 4-10 amino acids long, belonging to the family of atypical opioid peptides released during the sequential cleavage of hemoglobin protein. Hemorphins have been shown to exhibit diverse therapeutic effects in both human and animal models. However, the precise cellular and molecular mechanisms involved in such effects remain elusive. In this review, we summarize and propose potential mechanisms based on studies that investigated the biological activity of hemorphins of different lengths on multiple therapeutic targets. Special emphasis is given to molecular events related to renin-angiotensin system (RAS), opioid receptors and insulin-regulated aminopeptidase receptor (IRAP). This review provides a comprehensive coverage of the molecular mechanisms that underpin the therapeutic potential of hemorphins. Furthermore, it highlights the role of various hemorphin residues in pathological conditions, which could be explored further for therapeutic purposes.


Asunto(s)
Péptidos Opioides/fisiología , Péptidos Opioides/uso terapéutico , Animales , Humanos , Proteína Antagonista del Receptor de Interleucina 1/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Péptidos Opioides/química , Receptores Opioides/efectos de los fármacos , Sistema Renina-Angiotensina/efectos de los fármacos
4.
Int J Mol Sci ; 21(22)2020 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-33233481

RESUMEN

World Health Organization data suggest that stress, depression, and anxiety have a noticeable prevalence and are becoming some of the most common causes of disability in the Western world. Stress-related disorders are considered to be a challenge for the healthcare system with their great economic and social impact. The knowledge on these conditions is not very clear among many people, as a high proportion of patients do not respond to the currently available medications for targeting the monoaminergic system. In addition, the use of clinical drugs is also associated with various side effects such as vomiting, dizziness, sedation, nausea, constipation, and many more, which prevents their effective use. Therefore, opioid peptides derived from food sources are becoming one of the safe and natural alternatives because of their production from natural sources such as animals and plant proteins. The requirement for screening and considering dietary proteins as a source of bioactive peptides is highlighted to understand their potential roles in stress-related disorders as a part of a diet or as a drug complementing therapeutic prescription. In this review, we discussed current knowledge on opioid endogenous and exogenous peptides concentrating on their production, purification, and related studies. To fully understand their potential in stress-related conditions, either as a drug or as a therapeutic part of a diet prescription, the need to screen more dietary proteins as a source of novel opioid peptides is emphasized.


Asunto(s)
Ansiedad/prevención & control , Depresión/prevención & control , Péptidos Opioides/uso terapéutico , Trastornos de Estrés Traumático Agudo/prevención & control , Ansiedad/patología , Atención a la Salud , Depresión/patología , Humanos , Trastornos de Estrés Traumático Agudo/patología , Mundo Occidental
5.
Handb Exp Pharmacol ; 247: 277-299, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28315071

RESUMEN

Research of the opioid system and its composite receptors and ligands has revealed its promise as a potential therapy for neurodegenerative diseases such as stroke and Parkinson's Disease. In particular, delta opioid receptors (DORs) have been elucidated as a therapeutically distinguished subset of opioid receptors and a compelling target for novel intervention techniques. Research is progressively shedding light on the underlying mechanism of DORs and has revealed two mechanisms of DOR neuroprotection; DORs function to maintain ionic homeostasis and also to trigger endogenous neuroprotective pathways. Delta opioid agonists such as (D-Ala2, D-Leu5) enkephalin (DADLE) have been shown to promote neuronal survival and decrease apoptosis, resulting in a substantial amount of research for its application as a neurological therapeutic. Most notably, DADLE has demonstrated significant potential to reduce cell death following ischemic events. Current research is working to reveal the complex mechanisms of DADLE's neuroprotective properties. Ultimately, our knowledge of the DOR receptors and agonists has made the opioid system a promising target for therapeutic intervention in many neurological disorders.


Asunto(s)
Leucina Encefalina-2-Alanina/farmacología , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Péptidos Opioides/farmacología , Receptores Opioides delta/efectos de los fármacos , Accidente Cerebrovascular/tratamiento farmacológico , Animales , Leucina Encefalina-2-Alanina/uso terapéutico , Humanos , Enfermedades del Sistema Nervioso/fisiopatología , Péptidos Opioides/uso terapéutico , Accidente Cerebrovascular/fisiopatología
6.
Anesthesiology ; 124(3): 706-20, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26756519

RESUMEN

BACKGROUND: Opioids have long been regarded as the most effective drugs for the treatment of severe acute and chronic pain. Unfortunately, their therapeutic efficacy and clinical utility have been limited because of central and peripheral side effects. METHODS: To determine the therapeutic value of peripheral µ-opioid receptors as a target for neuropathic pain treatment, the authors examined the effects of dermorphin [D-Arg2, Lys4] (1-4) amide (DALDA), a hydrophilic, peripherally acting µ-opioid receptor agonist, in male and female rats with spinal nerve ligation-induced neuropathic pain. The authors also utilized behavioral, pharmacologic, electrophysiologic, and molecular biologic tools to characterize DALDA's possible mechanisms of action in male rats. RESULTS: DALDA, administered subcutaneously, had 70 times greater efficacy for inhibiting thermal (n = 8 to 11/group) than mechanical hypersensitivity (n = 6 to 8/group) in male rats. The pain inhibitory effects of DALDA on mechanical and heat hypersensitivity were abolished in animals pretreated with systemic methylnaltrexone (n = 7 to 9/group), a peripheral µ-opioid receptor antagonist. In the spinal wide-dynamic range neurons, systemic DALDA inhibited C-fiber-mediated, but not A-fiber-mediated, response in neuropathic male rats (n = 13). In primary sensory neurons, DALDA inhibited the capsaicin-induced [Ca2+] increase more than the ß-alanine-induced [Ca] increase (n = 300); capsaicin and ß-alanine activate subpopulations of neurons involved in the signaling of heat and mechanical pain, respectively. DALDA-treated rats (n = 5 to 8/group) did not exhibit motor deficits and locomotor impairment suggesting that it does not induce central side effects. CONCLUSIONS: These findings suggest that DALDA may represent a potential alternative to current opioid therapy for the treatment of neuropathic pain and is likely to be associated with minimal adverse effects.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Neuralgia/tratamiento farmacológico , Neuralgia/metabolismo , Péptidos Opioides/uso terapéutico , Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Animales , Femenino , Masculino , Péptidos Opioides/farmacología , Ratas , Ratas Sprague-Dawley
7.
Mol Pain ; 11: 2, 2015 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-25563474

RESUMEN

BACKGROUND: The treatment of spinal cord injury (SCI)-induced neuropathic pain presents a challenging healthcare problem. The lack of available robust pharmacological treatments underscores the need for novel therapeutic methods and approaches. Due to the complex character of neuropathic pain following SCI, therapies targeting multiple mechanisms may be a better choice for obtaining sufficient long-term pain relief. Previous studies in our lab showed analgesic effects using combinations of an NMDA antagonist peptide [Ser1]histogranin (SHG), and the mu-opioid peptides endomorphins (EMs), in several pain models. As an alternative to drug therapy, this study evaluated the analgesic potential of these peptides when delivered via gene therapy. RESULTS: Lentiviruses encoding SHG and EM-1 and EM-2 were intraspinally injected, either singly or in combination, into rats with clip compression SCI 2 weeks following injury. Treated animals showed significant reduction in mechanical and thermal hypersensitivity, compared to control groups injected with GFP vector only. The antinociceptive effects of individually injected components were modest, but the combination of EMs and SHG produced robust and sustained antinociception. The onset of the analgesic effects was observed between 1-5 weeks post-injection and sustained without decrement for at least 7 weeks. No adverse effects on locomotor function were observed. The involvement of SHG and EMs in the observed antinociception was confirmed by pharmacologic inhibition using intrathecal injection of either the opioid antagonist naloxone or an anti-SHG antibody. Immunohistochemical analysis showed the presence of SHG and EMs in the spinal cord of treated animals, and immunodot-blot analysis of CSF confirmed the presence of these peptides in injected animals. In a separate group of rats, delayed injection of viral vectors was performed in order to mimic a more likely clinical scenario. Comparable and sustained antinociceptive effects were observed in these animals using the SHG-EMs combination vectors compared to the group with early intervention. CONCLUSIONS: Findings from this study support the potential for direct gene therapy to provide a robust and sustained alleviation of chronic neuropathic pain following SCI. The combination strategy utilizing potent mu-opioid peptides with a naturally-derived NMDA antagonist may produce additive or synergistic analgesic effects without the tolerance development for long-term management of persistent pain.


Asunto(s)
Neuralgia/tratamiento farmacológico , Neuralgia/etiología , Péptidos Opioides/uso terapéutico , Proteínas/uso terapéutico , Traumatismos de la Médula Espinal/complicaciones , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Vectores Genéticos/fisiología , Humanos , Hiperalgesia/tratamiento farmacológico , Lentivirus/genética , Masculino , Neuroblastoma/patología , Neuropéptidos/biosíntesis , Neuropéptidos/uso terapéutico , Péptidos Opioides/biosíntesis , Péptidos Opioides/genética , Dimensión del Dolor , Umbral del Dolor/efectos de los fármacos , Proteínas/genética , Ratas , Ratas Sprague-Dawley , Recuperación de la Función/efectos de los fármacos
8.
Mol Pain ; 10: 10, 2014 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-24499354

RESUMEN

BACKGROUND: Leukocytes containing opioid peptides locally control inflammatory pain. In the early phase of complete Freund's adjuvant (CFA)-induced hind paw inflammation, formyl peptides (derived e.g. from Mycobacterium butyricum) trigger the release of opioid peptides from neutrophils contributing to tonic basal antinociception. In the later phase we hypothesized that toll-like-receptor-(TLR)-4 activation of monocytes/macrophages triggers opioid peptide release and thereby stimulates peripheral opioid-dependent antinociception. RESULTS: In Wistar rats with CFA hind paw inflammation in the later inflammatory phase (48-96 h) systemic leukocyte depletion by cyclophosphamide (CTX) or locally injected naloxone (NLX) further decreased mechanical and thermal nociceptive thresholds. In vitro ß-endorphin (ß-END) content increased during human monocyte differentiation as well as in anti-inflammatory CD14+CD16- or non-classical M2 macrophages. Monocytes expressing TLR4 dose-dependently released ß-END after stimulation with lipopolysaccharide (LPS) dependent on intracellular calcium. Despite TLR4 expression proinflammatory M1 and anti-inflammatory M2 macrophages only secreted opioid peptides in response to ionomycin, a calcium ionophore. Intraplantar injection of LPS as a TLR4 agonist into the inflamed paw elicited an immediate opioid- and dose-dependent antinociception, which was blocked by TAK-242, a small-molecule inhibitor of TLR4, or by peripheral applied NLX. In the later phase LPS lowered mechanical and thermal nociceptive thresholds. Furthermore, local peripheral TLR4 blockade worsened thermal and mechanical nociceptive pain thresholds in CFA inflammation. CONCLUSION: Endogenous opioids from monocytes/macrophages mediate endogenous antinociception in the late phase of inflammation. Peripheral TLR4 stimulation acts as a transient counter-regulatory mechanism for inflammatory pain in vivo, and increases the release of opioid peptides from monocytes in vitro. TLR4 antagonists as new treatments for sepsis and neuropathic pain might unexpectedly transiently enhance pain by impairing peripheral opioid analgesia.


Asunto(s)
Analgesia , Inflamación/tratamiento farmacológico , Péptidos Opioides/uso terapéutico , Receptor Toll-Like 4/metabolismo , Animales , Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , Adyuvante de Freund/administración & dosificación , Adyuvante de Freund/farmacología , Humanos , Hiperalgesia/complicaciones , Hiperalgesia/metabolismo , Hiperalgesia/patología , Inflamación/complicaciones , Inflamación/metabolismo , Inflamación/patología , Receptores de Lipopolisacáridos/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Masculino , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Nocicepción/efectos de los fármacos , Péptidos Opioides/farmacología , Ratas , Ratas Wistar , Receptores de IgG/metabolismo , Receptores Opioides/metabolismo , Receptor Toll-Like 2/metabolismo , betaendorfina/metabolismo
10.
Mol Neurobiol ; 61(8): 6089-6100, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38277118

RESUMEN

Cold injury or frostbite is a common medical condition that causes serious clinical complications including sensory abnormalities and chronic pain ultimately affecting overall well-being. Opioids are the first-choice drug for the treatment of frostbite-induced chronic pain; however, their notable side effects, including sedation, motor incoordination, respiratory depression, and drug addiction, present substantial obstacle to their clinical utility. To address this challenge, we have exploited peripheral mu-opioid receptors as potential target for the treatment of frostbite-induced chronic pain. In this study, we investigated the effect of dermorphin [D-Arg2, Lys4] (1-4) amide (DALDA), a peripheral mu-opioid receptor agonist, on frostbite injury and hypersensitivity induced by deep freeze magnet exposure in rats. Animals with frostbite injury displayed significant hypersensitivity to mechanical, thermal, and cold stimuli which was significant ameliorated on treatment with different doses of DALDA (1, 3, and 10 mg/kg) and ibuprofen (100 mg/kg). Further, molecular biology investigations unveiled heightened oxido-nitrosative stress, coupled with a notable upregulation in the expression of TRP channels (TRPA1, TRPV1, and TRPM8), glial cell activation, and neuroinflammation (TNF-α, IL-1ß) in the sciatic nerve, dorsal root ganglion (DRG), and spinal cord of frostbite-injured rats. Treatment with DALDA leads to substantial reduction in TRP channels, microglial activation, and suppression of the inflammatory cascade in the ipsilateral L4-L5 DRG and spinal cord of rats. Overall, findings from the present study suggest that activation of peripheral mu-opioid receptors mitigates chronic pain in rats by modulating the expression of TRP channels and suppressing glial cell activation and neuroinflammation.


Asunto(s)
Congelación de Extremidades , Microglía , Enfermedades Neuroinflamatorias , Péptidos Opioides , Ratas Sprague-Dawley , Animales , Masculino , Ratas , Congelación de Extremidades/tratamiento farmacológico , Congelación de Extremidades/complicaciones , Congelación de Extremidades/patología , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/patología , Hiperalgesia/metabolismo , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/metabolismo , Péptidos Opioides/metabolismo , Péptidos Opioides/farmacología , Péptidos Opioides/uso terapéutico , Dolor/tratamiento farmacológico , Dolor/metabolismo , Dolor/patología , Receptores Opioides mu/metabolismo , Receptores Opioides mu/agonistas , Canales de Potencial de Receptor Transitorio/metabolismo
11.
Invest New Drugs ; 31(4): 1066-70, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23275062

RESUMEN

Hepatoblastoma is the most common liver malignancy in children, typically diagnosed before age 2. The survival rate for hepatoblastoma has increased dramatically in the last 30 years, but the typical chemotherapeutic agents used for treatment are associated with significant toxicity. In this report, the authors present two cases of hepatoblastoma treated with surgical resection and a novel biotherapeutic regimen that included opioid growth factor (OGF). Case #1 is an infant diagnosed with a large mass on prenatal ultrasound. After subsequent diagnosis of hepatoblastoma, she was treated with one course of neoadjuvant chemotherapy at approximately 1 week of age. Following significant complications from the chemotherapy (neutropenic fever, pneumonia and sepsis), the patient's parents declined further chemotherapy, and the infant was treated with surgical resection and opioid growth factor (OGF)/low dose naltrexone (LDN). She is currently at close to 10 years disease-free survival. Case #2 is a child diagnosed with a liver mass on ultrasound at 20 months of age, later biopsy-proven to represent hepatoblastoma. Due to existing co-morbidities including autosomal recessive polycystic kidney disease and hypertension, and indications from the biopsy that the tumor might be insensitive to chemotherapy, the parents elected not to proceed with neoadjuvant chemotherapy. The patient was treated with surgical resection and OGF/LDN, and is currently at more than 5 years disease-free survival. This case series highlights the need for less toxic treatment options than conventional chemotherapy. Modulation of the OGF-OGF receptor axis represents a promising safe and therapeutic avenue for effective treatment of hepatoblastoma.


Asunto(s)
Encefalina Metionina/uso terapéutico , Hepatoblastoma/tratamiento farmacológico , Péptidos Opioides/uso terapéutico , Quimioterapia Adyuvante , Niño , Preescolar , Femenino , Hepatoblastoma/diagnóstico por imagen , Humanos , Lactante , Recién Nacido , Embarazo , Ultrasonografía
12.
Eur J Med Chem ; 258: 115608, 2023 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-37437352

RESUMEN

The compelling demand of a consummate analgesic medication without addiction is rising due to the clinical mistreatment. Additionally, the series of severe untoward effects usually deterred the utilization while coping with serious pain. As a possible turning point, we revealed that compound 14 is a dual agonist of mu opioid receptor (MOR) and nociceptin-orphanin FQ opioid peptide (NOP) receptor in this study. More importantly, compound 14 achieves pain relieving at very small doses, meanwhile, reduces several unwanted side effects such as constipation, reward, tolerance and withdrawal effects. Here, we evaluated the antinociception and side effects of this novel compound from wild type and humanized mice to further develop a safer prescription analgesic drug.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Receptores Opioides mu , Ratones , Animales , Receptores Opioides mu/agonistas , Receptores Opioides/agonistas , Receptor de Nociceptina , Péptidos Opioides/farmacología , Péptidos Opioides/uso terapéutico , Analgésicos Opioides/efectos adversos , Dolor/inducido químicamente , Dolor/tratamiento farmacológico , Analgésicos/efectos adversos , Nociceptina
13.
Clin Exp Pharmacol Physiol ; 39(5): 412-6, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22332877

RESUMEN

The opioid peptides have been implicated in peripheral antinociception induced by non-opioidergic compounds, including non-steroidal anti-inflammatory drugs and α(2) -adrenoceptor agonists. The aims of the present study were to investigate the possible peripheral antinociceptive effect of cafestol, a diterpene present in the oil derived from coffee beans, and to evaluate the involvement of opioid peptides in its effect. The rat paw pressure test was used to assess antinocipeptive effects. Hyperalgesia was induced by intraplantar injection of prostaglandin E(2) (2 µg/paw). All drugs were locally administered into the hind-paws of male Wistar rats. Intraplantar injection of cafestol (20, 40 and 80 µg) induced peripheral antinociception. The antinociceptive effect of cafestol was due to a local action because the higher dose (80 µg/paw) did not produce any effect in the contralateral paw. The opioid receptor antagonist naloxone (25, 50 and 100 µg/paw) prevented the action of cafestol (80 µg/paw), whereas the aminopeptidase inhibitor bestatin (400 µg/paw) potentiated the antinociceptive effect of cafestol (40 µg/paw). The results of the present study provide evidence that cafestol treatment has a peripheral antinociceptive effect and suggest that this effect is mediated by the release of endogenous opioids.


Asunto(s)
Café , Diterpenos/uso terapéutico , Hiperalgesia/tratamiento farmacológico , Péptidos Opioides/fisiología , Dimensión del Dolor/métodos , Animales , Café/química , Café/fisiología , Diterpenos/farmacología , Hiperalgesia/metabolismo , Hiperalgesia/patología , Masculino , Péptidos Opioides/uso terapéutico , Dimensión del Dolor/efectos de los fármacos , Ratas , Ratas Wistar
14.
Pharmacol Ther ; 231: 107982, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34480968

RESUMEN

The nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptor is a member of the opioid receptor superfamily with N/OFQ as its endogenous agonist. Wide expression of the NOP receptor and N/OFQ, both centrally and peripherally, and their ability to modulate several biological functions has led to development of NOP receptor modulators by pharmaceutical companies as therapeutics, based upon their efficacy in preclinical models of pain, anxiety, depression, Parkinson's disease, and substance abuse. Both posttraumatic stress disorder (PTSD) and traumatic brain injury (TBI) are debilitating conditions that significantly affect the quality of life of millions of people around the world. PTSD is often a consequence of TBI, and, especially for those deployed to, working and/or living in a war zone or are first responders, they are comorbid. PTSD and TBI share common symptoms, and negatively influence outcomes as comorbidities of the other. Unfortunately, a lack of effective therapies or therapeutic agents limits the long term quality of life for either TBI or PTSD patients. Ours, and other groups, demonstrated that PTSD and TBI preclinical models elicit changes in the N/OFQ-NOP receptor system, and that administration of NOP receptor ligands alleviated some of the neurobiological and behavioral changes induced by brain injury and/or traumatic stress exposure. Here we review the past and most recent progress on understanding the role of the N/OFQ-NOP receptor system in PTSD and TBI neurological and behavioral sequelae. There is still more to understand about this neuropeptide system in both PTSD and TBI, but current findings warrant further examination of the potential utility of NOP modulators as therapeutics for these disorders and their co-morbidities. We advocate the development of standards for common data elements (CDE) reporting for preclinical PTSD studies, similar to current preclinical TBI CDEs. That would provide for more standardized data collection and reporting to improve reproducibility, interpretation and data sharing across studies.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Calidad de Vida , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Humanos , Morbilidad , Péptidos Opioides/metabolismo , Péptidos Opioides/uso terapéutico , Reproducibilidad de los Resultados , Nociceptina
15.
Biomolecules ; 12(9)2022 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-36139079

RESUMEN

Despite various advantages, opioid peptides have been limited in their therapeutic uses due to the main drawbacks in metabolic stability, blood-brain barrier permeability, and bioavailability. Therefore, extensive studies have focused on overcoming the problems and optimizing the therapeutic potential. Currently, numerous peptide-based drugs are being marketed thanks to new synthetic strategies for optimizing metabolism and alternative routes of administration. This tutorial review briefly introduces the history and role of natural opioid peptides and highlights the key findings on their structure-activity relationships for the opioid receptors. It discusses details on opioid peptidomimetics applied to develop therapeutic candidates for the treatment of pain from the pharmacological and structural points of view. The main focus is the current status of various mimetic tools and the successful applications summarized in tables and figures.


Asunto(s)
Péptidos Opioides , Peptidomiméticos , Analgésicos Opioides/metabolismo , Analgésicos Opioides/farmacología , Analgésicos Opioides/uso terapéutico , Descubrimiento de Drogas , Péptidos Opioides/química , Péptidos Opioides/farmacología , Péptidos Opioides/uso terapéutico , Peptidomiméticos/farmacología , Peptidomiméticos/uso terapéutico , Receptores Opioides
16.
CNS Drugs ; 36(6): 617-632, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35616826

RESUMEN

Opioids are widely used in chronic pain management, despite major concerns about their risk of adverse events, particularly abuse, misuse, and respiratory depression from overdose. Multi-mechanistic opioids, such as tapentadol and buprenorphine, have been widely studied as a valid alternative to traditional opioids for their safer profile. Special interest was focused on the role of the nociceptin opioid peptide (NOP) receptor in terms of analgesia and improved tolerability. Nociceptin opioid peptide receptor agonists were shown to reinforce the antinociceptive effect of mu opioid receptor (MOR) agonists and modulate some of their adverse effects. Therefore, multi-mechanistic opioids involving both MOR and NOP receptor activation became a major field of pharmaceutical and clinical investigations. Buprenorphine was re-discovered in a new perspective, as an atypical analgesic and as a substitution therapy for opioid use disorders; and buprenorphine derivatives have been tested in animal models of nociceptive and neuropathic pain. Similarly, cebranopadol, a full MOR/NOP receptor agonist, has been clinically evaluated for its potent analgesic efficacy and better tolerability profile, compared with traditional opioids. This review overviews pharmacological mechanisms of the NOP receptor system, including its role in pain management and in the development of opioid tolerance. Clinical data on buprenorphine suggest its role as a safer alternative to traditional opioids, particularly in patients with non-cancer pain; while data on cebranopadol still require phase III study results to approve its introduction on the market. Other bifunctional MOR/NOP receptor ligands, such as BU08028, BU10038, and AT-121, are currently under pharmacological investigations and could represent promising analgesic agents for the future.


Asunto(s)
Analgésicos Opioides , Buprenorfina , Analgésicos Opioides/efectos adversos , Animales , Buprenorfina/farmacología , Buprenorfina/uso terapéutico , Tolerancia a Medicamentos , Humanos , Isoquinolinas , Naltrexona/análogos & derivados , Péptidos Opioides/uso terapéutico , Dolor/tratamiento farmacológico , Fenilpropionatos , Receptores Opioides mu/agonistas , Receptores Opioides mu/uso terapéutico , Nociceptina
17.
Headache ; 51(8): 1245-53, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21797854

RESUMEN

BACKGROUND: Alterations in the levels of nociceptin/orphanin FQ (N/OFQ) have been reported in patients with primary headaches, including migraines and cluster headaches. These clinical observations suggest that N/OFQ is involved in the pathogenesis of primary headaches. OBJECTIVES: The present study was conducted to determine the role of N/OFQ in the control of trigeminal nociception and cortical excitation. METHODS: Cortical spreading depression (CSD) was elicited in Wistar rats by cortical application of potassium chloride, and electrocorticograms were recorded. N/OFQ was administered via an intracisternal injection. The presence of CSD-evoked trigeminal nociception was determined with Fos and transient receptor potential vanilloid 1 (TRPV1) immunoreactivity. RESULTS: Nociceptin/orphanin FQ produced a biphasic effect on CSD generation, characterized by an initial attenuation followed by delayed potentiation. The amplitude of CSD waves were lower in the initial period but increased in the later period. The total number of CSD waves recorded in 1 hour was greater in the N/OFQ-treated group. Exposure to N/OFQ significantly increased the number of Fos-immunoreactive cells in the trigeminal nucleus caudalis and the number of TRPV1-immunoreactive cells in the trigeminal ganglia, indicating the enhancement of trigeminal nociception. CONCLUSION: These results indicate that N/OFQ can lead to biphasic effect characterized by an initial inhibition, and delay potentiation that eventually intensify CSD-evoked trigeminal nociception.


Asunto(s)
Depresión de Propagación Cortical/efectos de los fármacos , Nocicepción/fisiología , Péptidos Opioides/uso terapéutico , Nervio Trigémino/fisiopatología , Animales , Área Bajo la Curva , Depresión de Propagación Cortical/fisiología , Modelos Animales de Enfermedad , Electroencefalografía/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ratas , Ratas Wistar , Receptores Opioides/metabolismo , Canales Catiónicos TRPV/metabolismo , Núcleo Caudal del Trigémino/metabolismo , Ganglio del Trigémino/metabolismo , Neuralgia del Trigémino , Nociceptina
18.
Nat Med ; 9(8): 1003-8, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12894165

RESUMEN

The treatment of severe pain with opioids has thus far been limited by their unwanted central side effects. Recent research promises new approaches, including opioid analgesics acting outside the central nervous system, targeting of opioid peptide-containing immune cells to peripheral damaged tissue, and gene transfer to enhance opioid production at sites of injury.


Asunto(s)
Analgésicos Opioides/uso terapéutico , Péptidos Opioides/metabolismo , Manejo del Dolor , Dolor/metabolismo , Analgésicos Opioides/metabolismo , Animales , Movimiento Celular , Tolerancia a Medicamentos , Humanos , Ligandos , Neuronas Aferentes/metabolismo , Péptidos Opioides/uso terapéutico , Dolor/tratamiento farmacológico , Receptores Opioides/genética , Receptores Opioides/metabolismo , Transducción de Señal/fisiología
19.
Nihon Yakurigaku Zasshi ; 156(3): 139-144, 2021.
Artículo en Japonés | MEDLINE | ID: mdl-33952840

RESUMEN

After the identification of nociceptin/orphanin FQ (N/OFQ) peptide (NOP) and its cognate receptor, the unique functional profiles of the N/OFQ-NOP receptor system have been uncovered. NOP receptors are distributed in the key regions that regulate pain and reward processing in the central nervous system. In non-human primates (NHPs), activation of the NOP receptor causes antinociception and anti-hypersensitivity via spinal and supraspinal effects. Moreover, activation of the NOP receptor attenuates dopaminergic transmission and potentiates mu-opioid peptide (MOP) receptor-mediated analgesia. Here, we highlight the functional profiles of bifunctional NOP and MOP receptor agonists based on their promising effects for the treatment of pain and drug abuse. Bifunctional NOP/MOP receptor "partial" agonists, such as AT-121, BU08028, and BU10038, exert potent analgesic effects without MOP receptor-related side effects such as abuse liability, respiratory depression, physical dependence, and itching in NHPs. These novel NOP/MOP receptor agonists reduce rewarding and the reinforcing effects of abused drugs. Furthermore, a mixed NOP/opioid receptor "full" agonist, cebranopadol, is undergoing several clinical trials, and the therapeutic advantage of the coactivation of NOP and MOP receptors has also been confirmed in humans. Therefore, this class of drugs that coactivate NOP and MOP receptors proposes a wide therapeutic range with fewer side effects, indicating a greater potential for the development of novel safer opioid analgesics.


Asunto(s)
Analgésicos Opioides , Receptores Opioides , Analgésicos , Analgésicos Opioides/efectos adversos , Animales , Péptidos Opioides/uso terapéutico , Dolor/tratamiento farmacológico , Nociceptina
20.
Biomolecules ; 10(12)2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33333911

RESUMEN

In the last decade, natural-derived/-based biomolecules have continuously played an important role in novel drug discovery (as a prototype drug template) for potential chronic disease treatment [...].


Asunto(s)
Productos Biológicos/uso terapéutico , Enfermedad Crónica/terapia , Descubrimiento de Drogas , Animales , Humanos , Péptidos Opioides/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA