Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros










Intervalo de año de publicación
1.
J Neurosci ; 44(17)2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38438259

RESUMEN

Oxytocinergic transmission blocks nociception at the peripheral, spinal, and supraspinal levels through the oxytocin receptor (OTR). Indeed, a neuronal pathway from the hypothalamic paraventricular nucleus (PVN) to the spinal cord and trigeminal nucleus caudalis (Sp5c) has been described. Hence, although the trigeminocervical complex (TCC), an anatomical area spanning the Sp5c, C1, and C2 regions, plays a role in some pain disorders associated with craniofacial structures (e.g., migraine), the role of oxytocinergic transmission in modulating nociception at this level has been poorly explored. Hence, in vivo electrophysiological recordings of TCC wide dynamic range (WDR) cells sensitive to stimulation of the periorbital or meningeal region were performed in male Wistar rats. PVN electrical stimulation diminished the neuronal firing evoked by periorbital or meningeal electrical stimulation; this inhibition was reversed by OTR antagonists administered locally. Accordingly, neuronal projections (using Fluoro-Ruby) from the PVN to the WDR cells filled with Neurobiotin were observed. Moreover, colocalization between OTR and calcitonin gene-related peptide (CGRP) or OTR and GABA was found near Neurobiotin-filled WDR cells. Retrograde neuronal tracers deposited at the meningeal (True-Blue, TB) and infraorbital nerves (Fluoro-Gold, FG) showed that at the trigeminal ganglion (TG), some cells were immunopositive to both fluorophores, suggesting that some TG cells send projections via the V1 and V2 trigeminal branches. Together, these data may imply that endogenous oxytocinergic transmission inhibits the nociceptive activity of second-order neurons via OTR activation in CGRPergic (primary afferent fibers) and GABAergic cells.


Asunto(s)
Estimulación Eléctrica , Oxitocina , Núcleo Hipotalámico Paraventricular , Ratas Wistar , Receptores de Oxitocina , Transmisión Sináptica , Animales , Masculino , Núcleo Hipotalámico Paraventricular/fisiología , Núcleo Hipotalámico Paraventricular/metabolismo , Oxitocina/metabolismo , Oxitocina/análogos & derivados , Ratas , Receptores de Oxitocina/metabolismo , Receptores de Oxitocina/antagonistas & inhibidores , Transmisión Sináptica/fisiología , Nociceptores/fisiología , Nociceptores/metabolismo , Nocicepción/fisiología , Potenciales de Acción/fisiología , Potenciales de Acción/efectos de los fármacos , Meninges/fisiología , Inhibición Neural/fisiología
2.
Exp Neurol ; 363: 114383, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36921751

RESUMEN

Oxytocin receptor (OTR) activation at the spinal level produces antinociception. Some data suggest that central OTR activation enhances social interaction via an increase of endocannabinoids (eCB), but we do not know if this could occur at the spinal level, modulating pain transmission. Considering that oxytocin via OTR stimulates diacylglycerol formation, a key intermediate in synthesizing 2-arachidonylglycerol (2-AG), an eCB molecule, we sought to test the role of the eCB system on the spinal oxytocin-induced antinociception. Behavioral and electrophysiological experiments were conducted in naïve and formalin-treated (to induce long-term mechanical hypersensitivity) male Wistar rats. Intrathecal RHC 80267 injections, an inhibitor of the enzyme diacylglycerol lipase (thus, decreasing 2-AG formation), produces transient mechanical hypersensitivity, an effect unaltered by oxytocin but reversed by gabapentin. Similarly, in in vivo extracellular recordings of naïve spinal wide dynamic range cells, juxtacellular picoinjection of RHC 80267 increases the firing of nociceptive Aδ-, C-fibers, and post-discharge, an effect unaltered by oxytocin. Interestingly, in sensitized rats, oxytocin picoinjection reverses the RHC 80627-induced hyperactivity of Aδ-fibers (but not C- or post-discharge activity). In contrast, a sub-effective dose of JZL184 (a monoacylglycerol lipase inhibitor, thus favoring 2-AG levels), which does not have per se an antinociceptive effect in the formalin-induced hypernociception, the oxytocin-induced antinociception is boosted. Similarly, electrophysiological experiments suggest that juxtacellular JZL184 diminishes the neuronal firing of nociceptive fibers, and co-injection with oxytocin prolongs and enhances the antinociceptive effect. These data may imply that 2-AG formation may play a role in the spinal antinociception induced by oxytocin.


Asunto(s)
Endocannabinoides , Oxitocina , Ratas , Masculino , Animales , Humanos , Oxitocina/farmacología , Ratas Wistar , Cuidados Posteriores , Alta del Paciente , Receptores de Oxitocina , Fibras Nerviosas Amielínicas , Analgésicos/farmacología , Formaldehído
3.
Front Pharmacol ; 13: 1023611, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36506544

RESUMEN

Spinal α2-adrenoceptor induces analgesia by neuronal inhibition of primary afferent fibers. This family receptor coupled to G i/o proteins can be subdivided into three functional subtypes: α2A, α2B, and α2C-adrenoceptors, and current evidence on spinal analgesia supports the relevance of α2A and seems to exclude the role of α2B, but the functional contribution of α2C-adrenoceptors remains elusive. The present study was designed to pharmacologically dissect the contribution of spinal α2-adrenoceptor subtypes modulating tonic or acute peripheral nociception. Using male Wistar rats, we analyzed the effect of spinal clonidine (a non-selective α2A/α2B/α2C-adrenoceptor agonist) and/or selective subtype α2-adrenoceptor antagonists on: 1) tonic nociception induced by subcutaneous formalin (flinching behavior) or 2) acute nociception induced by peripheral electrical stimulus in in vivo extracellular recordings of spinal dorsal horn second-order wide dynamic range (WDR) neurons. Clonidine inhibited the nocifensive behavior induced by formalin, an effect blocked by BRL 44408 (α2A-adrenoceptor antagonist) but not by imiloxan (α2B-adrenoceptor antagonist) or JP 1302 (α2C-adrenoceptor antagonist). Similarly, spinal BRL 44408 reversed the clonidine-induced inhibition of nociceptive WDR activity. Interestingly, spinal JP 1302 per se produced behavioral antinociception (an effect blocked by bicuculline, a preferent GABAA channel blocker), but no correlation was found with the electrophysiological experiments. These data imply that, at the spinal level, 1) presynaptic α2A-adrenoceptor activation produces antinociception during acute or tonic nociceptive stimuli; and 2) under tonic nociceptive (inflammatory) input, spinal α2C-adrenoceptors are pronociceptive, probably by the inactivation of GABAergic transmission. This result supports a differential role of α2A and α2C-adrenoceptors modulating nociception.

4.
Eur J Pain ; 26(4): 796-810, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34978727

RESUMEN

BACKGROUND: Sex plays a crucial role in pain processing and response to analgesic drugs. Indeed, spinal glia seems to be significant in the sexual dimorphism observed in the above effects. Recently, studies have associated oxytocin with antinociceptive effects, but these have been mainly performed in male animals; consequently, the influence of sex has been poorly explored. METHODS: Using a model of spinal nociception that produces pain through activation of the spinal glia, that is, intrathecal (i.t.) lipopolysaccharide (LPS) injection, we analysed the changes in the analgesic response to i.t. oxytocin in female and male rats by behavioural (punctate mechanical hypersensitivity), electrophysiological (unitary extracellular recordings of wide dynamic range [WDR] cells) and molecular biology (real-time PCR of proinflammatory genes) experiments. RESULTS: We found that LPS-induced hypersensitivity was longer in female (>96 h) than in male (≈4 h) rats. Besides, spinal oxytocin preferentially prevents the LPS-induced hypersensitivity in male rather than female rats. Indeed, LPS increases the spinal neuronal-evoked activity associated with the activation of peripheral Aδ- and C-fibres and post-discharge in males, whereas only C-fibre discharge was enhanced in females. The electrophysiological data correlate with the fact that spinal oxytocin only prevented TNF-α and IL-1ß synthesis in male rats. CONCLUSIONS: Therefore, these data suggest that oxytocin-mediated analgesia depends on a sexual dimorphism involving activation of the spinal glia. These results reinforced the idea that different strategies are required to treat pain in men and women, and that oxytocin could be used preferentially to treat pain with a significant inflammatory component in men. SIGNIFICANCE STATEMENT: Oxytocin is a molecule that emerges as a potent analgesic in preclinical and clinical studies. We investigated the contribution of glia to the response of oxytocin-induced analgesia and how sex influences in this response show that different strategies are required to treat pain in men and women, and that oxytocin could be used preferentially to treat pain with a significant inflammatory component in men.


Asunto(s)
Cuidados Posteriores , Oxitocina , Analgésicos/farmacología , Analgésicos/uso terapéutico , Animales , Femenino , Humanos , Masculino , Fibras Nerviosas Amielínicas , Neuroglía , Oxitocina/farmacología , Alta del Paciente , Ratas , Médula Espinal
5.
Neuroscience ; 475: 117-126, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34530103

RESUMEN

Oxytocin is a hypothalamic neuropeptide involved in the inhibition of nociception transmission at spinal dorsal horn (SDH) level (the first station where the incoming peripheral signals is modulated). Electrophysiological, behavioral, and pharmacological data strongly support the role of this neuropeptide and its receptor (the oxytocin receptor, OTR) as a key endogenous molecule with analgesic properties. Briefly, current data showed that oxytocin release from the hypothalamus induces OTR activation at the SDH, inducing selective inhibition of the nociceptive Aδ- and C-fibers (probably peptidergic) activity, but not the activity of proprioceptive fibers (i.e. Aß-fibers). The above inhibition could be a direct presynaptic mechanism, or a mechanism mediated by GABAergic interneurons. However, the exact anatomical localization of oxytocin and OTR remains unclear. In this context, the present study set out to analyze the role of OTRs, GABAergic cells and CGRP fibers in the SDH in rats by using electron microscopy. Ultrastructural analyses of the SDH tissue show that: (i) oxytocin and OTR are found in asymmetrical synapsis; (ii) OTR is found in GABAergic interneurons (near unmyelinated fibers), CGRPergic fibers and glial cells; (iii) whereas oxytocin is present in supraspinal descending projection fibers. These anatomical data strongly support the notion that oxytocin released at the SDH could presynaptically inhibit the nociceptive input from the peripheral primary afferent fibers. This inhibitory action could be direct or use a GABA interneuron. Furthermore, our findings that OTR is exhibited in glial tissue at the SDH requires further exploration in nociception assays.


Asunto(s)
Nocicepción , Receptores de Oxitocina , Animales , Fibras Nerviosas Amielínicas , Oxitocina , Células del Asta Posterior , Ratas , Asta Dorsal de la Médula Espinal
6.
ACS Chem Neurosci ; 12(16): 3140-3147, 2021 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-34342984

RESUMEN

Behavioral and electrophysiological data show that at the spinal level, oxytocin inhibits pain transmission by activation of oxytocin receptors (OTRs). Canonically, OTRs are coupled to Gq proteins, which induce a rise of intracellular Ca2+ by activating the phospholipase C (PLC). However, in vitro data showed that OTRs cause a plethora of intracellular events, some related to the activation of Gi proteins. Using a behavioral approach, we analyzed the main in vivo intracellular pathway elicited by spinal oxytocin during a peripheral inflammatory/persistent nociceptive stimulus. Intrathecal oxytocin reduces early (number of flinches) and late (mechanical allodynia) formalin-induced nociception, an effect abolished by the OTR antagonist (L-368,899). Furthermore, the antinociception observed during the early phase (acute inflammatory) was also reverted by U-73122 (PLC inhibitor) but not by pertussis toxin (Gαi/o protein inhibitor) or gallein (Gßγ subunit inhibitor). In contrast, the late oxytocin-induced behavioral analgesia was blocked by pertussis and gallein but not by U-73122. Since oxytocin's effects during the early phase were also antagonized by Nω-nitro-l-arginine methyl ester, ODQ, or glibenclamide (inhibitors of nitric oxide synthase [NOS], soluble guanylyl cyclase [GC], and K+ATP channels, respectively), the role of two differential pathways elicited by oxytocin is supported. Hence, we showed in in vivo experiments that oxytocin recruits two differential spinal intracellular pathways mediated by Gq (PLC/NOS/GC/K+ATP) or Gi proteins during a peripheral nociceptive stimulus.


Asunto(s)
Oxitocina , Receptores de Oxitocina , Animales , Disección , Nocicepción , Oxitocina/farmacología , Ratas , Ratas Wistar
7.
Neuroscience ; 458: 256-270, 2021 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-33465410

RESUMEN

Nociception is the neuronal process of encoding noxious stimuli and could be modulated at peripheral, spinal, brainstem, and cortical levels. At cortical levels, several areas including the anterior cingulate cortex (ACC), prefrontal cortex (PFC), ventrolateral orbital cortex (VLO), insular cortex (IC), motor cortex (MC), and somatosensory cortices are involved in nociception modulation through two main mechanisms: (i) a descending modulatory effect at spinal level by direct corticospinal projections or mostly by activation of brainstem structures (i.e. periaqueductal grey matter (PAG), locus coeruleus (LC), the nucleus of raphe (RM) and rostroventral medulla (RVM)); and by (ii) cortico-cortical or cortico-subcortical interactions. This review summarizes evidence related to the participation of the aforementioned cortical areas in nociception modulation and different neurotransmitters or neuromodulators that have been studied in each area. Besides, we point out the importance of considering intracortical neuronal populations and receptors expression, as well as, nociception-induced cortical changes, both functional and connectional, to better understand this modulatory effect. Finally, we discuss the possible mechanisms that could potentiate the use of cortical stimulation as a promising procedure in pain alleviation.


Asunto(s)
Nocicepción , Sustancia Gris Periacueductal , Humanos , Locus Coeruleus , Bulbo Raquídeo , Vías Nerviosas , Dolor
8.
J Neurosci Methods ; 350: 109048, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33359224

RESUMEN

BACKGROUND: The CLARITY technique enables researchers to visualize different neuronal connections along the nervous system including the somatosensory system. NEW METHOD: The present work describes the antero-lateral and dorsal column pathways until the thalamic and cortical stations, as well as descending oxytocinergic and vasopressinergic innervations by means of combined CLARITY, neuronal tracing, and immunofluorescence techniques. We used male Sprague-Dawley rats of 13, 30, and 60 days. RESULTS: The main results are as follows: A) CLARITY is a reliable technique that can be combined with fluorescent neuronal tracers and immunofluorescence techniques without major procedure modifications; B) at spinal level, some primary afferent fibers were labeled by CGRP, as well as the presence of neuronal populations that simultaneously project to the gracile and ventral posterolateral thalamic nuclei; C) corticothalamic connections were visible when retrograde tracers were injected at thalamic level; D) oxytocin receptors were expressed in the spinal dorsal horn by GABAergic-positive neurons, reinforcing previous outcomes about the possible mechanism for oxytocin blocking the primary afferent sensory input. COMPARISON WITH EXISTING METHODS AND CONCLUSIONS: The CLARITY technique lets us observe in a transparent way the entire processed tissue compared with classical histological methods. CLARITY is a potentially useful tool to describe neuroanatomical structures and their neurochemical stratus.


Asunto(s)
Neuronas , Núcleos Talámicos Ventrales , Animales , Axones , Técnica del Anticuerpo Fluorescente , Masculino , Ratas , Ratas Sprague-Dawley
9.
Front Pharmacol ; 11: 581544, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33071793

RESUMEN

The first few days post-surgery, patients experience intense pain, hypersensitivity and consequently tend to have minor locomotor activity to avoid pain. Certainly, injury to peripheral tissues produces pain and increases sensitivity to painful (hyperalgesia) and non-painful (allodynia) stimuli. In this regard, preemptive pharmacological treatments to avoid or diminish pain after surgery are relevant. Recent data suggest that the neuropeptide oxytocin when given at spinal cord level could be a molecule with potential preemptive analgesic effects, but this hypothesis has not been properly tested. Using a validated postoperative pain model (i.e. plantar incision), we evaluated in male Wistar rats the potential preemptive antinociceptive effects of intrathecal oxytocin administration measuring tactile hypersensitivity (across 8 days) and spontaneous motor activity (across 3 days). Hypersensitivity was evaluated using von Frey filaments, whereas spontaneous activity (total distance, vertical activity episodes, and time spent in the center of the box) was assessed in real time using a semiautomated open-field system. Under these conditions, we found that animals pretreated with spinal oxytocin before plantar incision showed a diminution of hypersensitivity and an improvement of spontaneous behavior (particularly total distance and vertical activity episodes). This report provides a basis for addressing the therapeutic relevance of oxytocin as a potential preemptive analgesic molecule.

10.
Cell Calcium ; 90: 102246, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32590238

RESUMEN

Recently, oxytocin (OT) has been studied as a potential modulator of endogenous analgesia by acting upon pain circuits at the spinal cord and supraspinal levels. Yet the detailed action mechanisms of OT are still undetermined. The present study aimed to evaluate the action of OT in the spinal cord dorsal horn network under nociceptive-like conditions induced by the activation of the N-methyl-d-aspartate (NMDA) receptor and formalin injection, using calcium imaging techniques. Results demonstrate that the spontaneous Ca2+-dependent activity of the dorsal horn cells was scarce, and the coactivity of cells was mainly absent. When NMDA was applied, high rates of activity and coactivity occurred in the dorsal horn cells; these rates of high activity mimicked the activity dynamics evoked by a neuropathic pain condition. In addition, although OT treatment increased activity rates, it was also capable of disrupting the conformation of coordinated activity previously consolidated by NMDA treatment, without showing any effect by itself. Altogether, our results suggest that OT globally prevents the formation of coordinated patterns previously generated by nociceptive-like conditions on dorsal horn cells by NMDA application, which supports previous evidence showing that OT represents a potential therapeutic alternative for the treatment of chronic neuropathic pain.


Asunto(s)
Red Nerviosa/patología , Neuralgia/patología , Neuronas/patología , Oxitocina/farmacología , Asta Dorsal de la Médula Espinal/patología , Animales , Animales Recién Nacidos , Calcio/metabolismo , Femenino , Formaldehído/administración & dosificación , Masculino , N-Metilaspartato/farmacología , Red Nerviosa/efectos de los fármacos , Neuronas/efectos de los fármacos
11.
J Neurosci ; 40(29): 5669-5680, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32571836

RESUMEN

The rostral agranular insular cortex (RAIC) is a relevant structure in nociception. Indeed, recruitment of GABAergic activity in RAIC promotes the disinhibition of the locus ceruleus, which in turn inhibits (by noradrenergic action) the peripheral nociceptive input at the spinal cord level. In this regard, at the cortical level, oxytocin can modulate the GABAergic transmission; consequently, an interaction modulating nociception could exist between oxytocin and GABA at RAIC. Here, we tested in male Wistar rats the effect of oxytocin microinjection into RAIC during an inflammatory (by subcutaneous peripheral injection of formalin) nociceptive input. Oxytocin microinjection produces a diminution of (1) flinches induced by formalin and (2) spontaneous firing of spinal wide dynamic range cells. The above antinociceptive effect was abolished by microinjection (at RAIC) of the following: (1) L-368899 (an oxytocin receptor [OTR] antagonist) or by (2) bicuculline (a preferent GABAA receptor blocker), suggesting a GABAergic activation induced by OTR. Since intrathecal injection of an α2A-adrenoceptor antagonist (BRL 44408) partially reversed the oxytocin effect, a descending noradrenergic antinociception is suggested. Further, injection of L-368899 per se induces a pronociceptive behavioral effect, suggesting a tonic endogenous oxytocin release during inflammatory nociceptive input. Accordingly, we found bilateral projections from the paraventricular nucleus of the hypothalamus (PVN) to RAIC. Some of the PVN-projecting cells are oxytocinergic and destinate GABAergic and OTR-expressing cells inside RAIC. Aside from the direct anatomic link between PVN and RAIC, our findings provide evidence about the role of oxytocinergic mechanisms modulating the pain process at the RAIC level.SIGNIFICANCE STATEMENT Oxytocin is a neuropeptide involved in several functions ranging from lactation to social attachment. Over the years, the role of this molecule in pain processing has emerged, showing that, at the spinal level, oxytocin blocks pain transmission. The present work suggests that oxytocin also modulates pain at the cortical insular level by favoring cortical GABAergic transmission and activating descending spinal noradrenergic mechanisms. Indeed, we show that the paraventricular hypothalamicnucleus sends direct oxytocinergic projections to the rostral agranular insular cortex on GABAergic and oxytocin receptor-expressing neurons. Together, our data support the notion that the oxytocinergic system could act as an orchestrator of pain modulation.


Asunto(s)
Corteza Cerebral/fisiología , Inflamación/fisiopatología , Neuronas/fisiología , Nocicepción/fisiología , Oxitocina/fisiología , Animales , Corteza Cerebral/citología , Corteza Cerebral/efectos de los fármacos , Formaldehído/administración & dosificación , Neuronas GABAérgicas/fisiología , Inflamación/inducido químicamente , Masculino , Vías Nerviosas/citología , Vías Nerviosas/fisiología , Nocicepción/efectos de los fármacos , Oxitocina/administración & dosificación , Núcleo Hipotalámico Paraventricular/citología , Núcleo Hipotalámico Paraventricular/fisiología , Ratas Wistar
12.
Exp Neurol ; 323: 113079, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31678349

RESUMEN

Migraine is a complex brain disorder that involves abnormal activation of the trigeminocervical complex (TCC). Since an increase of oxytocin concentration has been found in cerebrospinal fluid in migrainous patients and intranasal oxytocin seems to relieve migrainous pain, some studies suggest that the hypothalamic neuropeptide oxytocin may play a role in migraine pathophysiology. However, it remains unknown whether oxytocin can interact with the trigeminovascular system at TCC level. The present study was designed to test the above hypothesis in a well-established electrophysiological model of migraine. Using anesthetized rats, we evaluated the effect of oxytocin on TCC neuronal activity in response to dural nociceptive trigeminovascular activation. We found that spinal oxytocin significantly reduced TCC neuronal firing evoked by meningeal electrical stimulation. Furthermore, pretreatment with L-368,899 (a selective oxytocin receptor antagonist, OTR) abolished the oxytocin-induced inhibition of trigeminovascular neuronal responses. This study provides the first direct evidence that oxytocin, probably by OTR activation at TCC level inhibited dural nociceptive-evoked action potential in this complex. Thus, targeting OTR at TCC could represent a new avenue to treat migraine.


Asunto(s)
Duramadre/fisiología , Nocicepción/fisiología , Oxitocina/metabolismo , Transmisión Sináptica/fisiología , Núcleos del Trigémino/fisiología , Potenciales de Acción/fisiología , Animales , Estimulación Eléctrica , Trastornos Migrañosos/metabolismo , Trastornos Migrañosos/fisiopatología , Ratas , Ratas Wistar
13.
Exp Brain Res ; 237(11): 2995-3010, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31515587

RESUMEN

The search for new ligands to treat neuropathic pain remains a challenge. Recently, oxytocin has emerged as an interesting molecule modulating nociception at central and peripheral levels, but no attempt has been made to evaluate the effect of recurrent oxytocin administration in neuropathic pain. Using male Wistar rats with spinal nerve ligation, we evaluated the effects of recurrent spinal (1 nmol; given by lumbar puncture) or peripheral (31 nmol; given by intraplantar injection in the ipsilateral paw to spinal nerve ligation) oxytocin administration on pain-like behavior in several nociceptive tests (tactile allodynia and thermal and mechanical hyperalgesia) on different days. Furthermore, we used an electrophysiological approach to analyze the effect of spinal 1 nmol oxytocin on the activity of spinal dorsal horn wide dynamic range cells. In neuropathic rats, spinal or peripheral oxytocin partially restored the nociceptive threshold measured with the von Frey filaments (tactile allodynia), Hargreaves (thermal hyperalgesia) and Randall-Selitto (mechanical hyperalgesia) tests for 12 days. These results agree with electrophysiological data showing that spinal oxytocin diminishes the neuronal firing of the WDR neurons evoked by peripheral stimulation. This effect was associated with a decline in the activity of primary afferent Aδ- and C-fibers. The above findings show that repeated spinal or peripheral oxytocin administration attenuates the pain-like behavior in a well-established model of neuropathic pain. This study provides a basis for addressing the therapeutic relevance of oxytocin in chronic pain conditions.


Asunto(s)
Conducta Animal/efectos de los fármacos , Hiperalgesia/tratamiento farmacológico , Neuralgia/tratamiento farmacológico , Nocicepción/efectos de los fármacos , Oxitocina/farmacología , Animales , Modelos Animales de Enfermedad , Inyecciones Intramusculares , Inyecciones Espinales , Masculino , Oxitocina/administración & dosificación , Ratas , Ratas Wistar
14.
Eur J Pain ; 23(8): 1519-1526, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31116891

RESUMEN

BACKGROUND: Preemptive analgesia encompasses different perioperative interventions that have the final aim of decreasing postoperative pain and improving recovery. Recently, peripheral analgesic effects of oxytocinergic modulation have been suggested. In this regard, we tested the potential analgesic effects of subcutaneous oxytocin (OT) infiltration in patients submitted to laparoscopic cholecystectomy. METHODS: Thirty patients with similar general characteristics and medical physical conditions were evaluated. The patients were assigned by simple random selection to one of three groups: (a) OT group (n = 10), which received preincisional subcutaneous OT (4 µg/4 ml saline) in the surgical sites for trocar placements; (b) Lidocaine group (n = 10), which received subcutaneous lidocaine 1% (4 ml) in the surgical sites; and (c) Control group (n = 10), which did not receive any treatment. Then we measured the effect of those treatments on the hemodynamic variations produced as responses to the surgical incisions and trocar insertions (open port placements using the Hasson technique). Moreover, we assessed the intensity of postoperative pain with the visual analogue scale during recovery and 24 hr after surgery. RESULTS: Hemodynamic parameters were stable in both intervention groups (subcutaneous OT and lidocaine) during the surgical incisions and trocar placements, whereas a most likely sympathetic activation due to trocar insertions (open port placements) was not blunted in the placebo group. Furthermore, postoperative pain was diminished in both OT and lidocaine groups when compared to the control group. CONCLUSIONS: Preincisional subcutaneous OT administration reduced the hemodynamic response produced by the latter. Furthermore, OT also diminished postoperative pain.


Asunto(s)
Hemodinámica/efectos de los fármacos , Laparoscopía , Lidocaína/administración & dosificación , Oxitocina/uso terapéutico , Dolor Postoperatorio/tratamiento farmacológico , Herida Quirúrgica/tratamiento farmacológico , Adulto , Anestesia General , Anestésicos Locales/administración & dosificación , Femenino , Humanos , Masculino , Persona de Mediana Edad , Dimensión del Dolor , Distribución Aleatoria
15.
Neurosci Lett ; 696: 20-27, 2019 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-30543847

RESUMEN

Neuronal activity can be modulated by endogenous control mechanisms that either facilitate or suppress it. With this idea in mind, we attempted to evaluate and correlate spinal neuronal activity with the amplitude of corticogram (ECoG) event related potentials (ERP) in the presence of nociceptive stimulation in rats. We evaluated the ERP in response to noxious stimuli, endogenous analgesic actions, different frequencies, and heterotopic nociceptive stimulation, as well as in conjunction with recordings from neurons in the spinal cord that are activated by noxious stimuli. Computational tasks enabled us to establish correlations between the amplitude of ERP and neuronal firing of cells in the spinal dorsal horn. Our results show that the ERP amplitude could be modified by previous activity in the cerebral cortex, but the activity in the spinal cord did not change. Previous activity could originate spontaneously or could be driven by sensory stimulation. A recurrent inhibitory cortical action is proposed that could explain the suppression of pain perception during electrical or magnetic transcranial stimulation, as well as during heterotopic stimulation. This study aims to uncover a local recurrent inhibitory cortical action that could modify the sensory information.


Asunto(s)
Corteza Cerebral/fisiología , Potenciales Evocados/fisiología , Dolor/fisiopatología , Médula Espinal/fisiología , Potenciales de Acción/fisiología , Animales , Estimulación Eléctrica/métodos , Neuronas/fisiología , Ratas Sprague-Dawley , Ratas Wistar
16.
Neurosci Lett ; 685: 124-130, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30130553

RESUMEN

Hypothalamic paraventricular nucleus (PVN) projections to the spinal dorsal horn (SDH) are related to antinociception. Several neuropeptides from this nucleus could be released to the spinal cord after nociceptive stimuli. Indeed, it has been shown that enkephalins, oxytocin and vasopressin could be released at this level. Although the antinociceptive effects of these neuropeptides are well studied, little is known about the potential interaction between these molecules. In this study, we provide anatomical evidence of the interaction between oxytocin (OT), vasopressin (AVP), dynorphin (DYN) and enkephalin (ENK) along the PVN projections to the spinal dorsal horn at L3 level. A retrograde tracer (True Blue®) microinjected at L3 in the SDH and immunofluorescence with antibodies against OT, AVP, DYN and ENK were used. The experiments showed different levels of peptide immunoreactivity distribution along the rostro-caudal area of the PVN. A high percentage of co-localizations between two of the peptides (OT-AVP, OT-DYN, AVP-ENK, DYN-ENK) were present along the PVN. The following co-localizations occupied 4.76-9.62% of the total PVN area. PVN projections to the SDH at L3 level showed similar results. Our results show that different antinociceptive peptides may be interacting with each other to evoke PVN antinociceptive effects as part of the endogenous system of nociceptive modulation.


Asunto(s)
Encefalinas/farmacología , Oxitocina/farmacología , Núcleo Hipotalámico Paraventricular/efectos de los fármacos , Médula Espinal/efectos de los fármacos , Vasopresinas/farmacología , Animales , Dinorfinas/farmacología , Encefalinas/metabolismo , Masculino , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Nocicepción/efectos de los fármacos , Núcleo Hipotalámico Paraventricular/metabolismo , Ratas Wistar , Médula Espinal/metabolismo , Asta Dorsal de la Médula Espinal/efectos de los fármacos , Asta Dorsal de la Médula Espinal/metabolismo
17.
Neuropharmacology ; 129: 109-117, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29169960

RESUMEN

The medullary dorsal horn (MDH or Sp5c/C1 region) plays a key role modulating the nociceptive input arriving from craniofacial structures. Some reports suggest that oxytocin could play a role modulating the nociceptive input at the MDH level, but no study has properly tested this hypothesis. Using an electrophysiological and pharmacological approach, the present study aimed to determine the effect of oxytocin on the nociceptive signaling in the MDH and the receptor involved. In sevoflurane, anesthetized rats, we performed electrophysiological unitary recordings of second order neurons at the MDH region responding to peripheral nociceptive-evoked responses of the first branch (V1; ophthalmic) of the trigeminal nerve. Under this condition, we constructed dose-response curves analyzing the effect of local spinal oxytocin (0.2-20 nmol) on MDH nociceptive neuronal firing. Furthermore, we tested the role of oxytocin receptors (OTR) or vasopressin V1A receptors (V1AR) involved in the oxytocin effects. Oxytocin dose-dependently inhibits the peripheral-evoked activity in nociceptive MDH neurotransmission. This inhibition is associated with a blockade of neuronal activity of Aδ- and C-fibers. Since this antinociception was abolished by pretreatment (in the MDH) with the potent and selective OTR antagonist (L-368,899; 20 nmol) and remained unaffected after the V1AR antagonist (SR49059; 20 nmol or 200 nmol), the role of OTR is implied. This electrophysiological study demonstrates that oxytocin inhibits the peripheral-evoked neuronal activity at MDH, through OTR activation. Thus, OTR may represent a new potential drug target to treat craniofacial nociceptive dysfunction in the MDH.


Asunto(s)
Nociceptores/efectos de los fármacos , Oxitócicos/farmacología , Oxitocina/farmacología , Receptores de Oxitocina/metabolismo , Receptores de Vasopresinas/metabolismo , Asta Dorsal de la Médula Espinal/citología , Potenciales de Acción/efectos de los fármacos , Análisis de Varianza , Animales , Antagonistas de los Receptores de Hormonas Antidiuréticas/farmacología , Canfanos/farmacología , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica , Indoles/farmacología , Masculino , Fibras Nerviosas/fisiología , Oxitocina/antagonistas & inhibidores , Piperazinas/farmacología , Pirrolidinas/farmacología , Ratas , Ratas Wistar
18.
Pain ; 158(11): 2117-2128, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28731982

RESUMEN

Oxytocin (OT) has emerged as a mediator of endogenous analgesia in behavioral and electrophysiological experiments. In fact, OT receptors (OTRs) in the spinal dorsal horn participate in a selective inhibition of the neuronal activity mediated by Aδ and C fibers but not Aß fibers. This study shows that OTRs are expressed in the terminal nerve endings and are able to inhibit nociceptive neuronal firing. Indeed, local peripheral OT blocked the first sensorial activity of Aδ and C fibers recorded in the spinal cord neurons. Furthermore, using the formalin behavioral nociceptive test, we demonstrated that only ipsilateral OTR activation inhibits pain behavior. Our data are reinforced by the fact that the OTR protein is expressed in the sciatic nerve. Consistent with this, immunofluorescence of primary afferent fibers suggest that OTRs could be located in nociceptive-specific terminals of the skin. Taken together, our results suggest that OTRs could be found in nociceptive terminals and that on activation they are able to inhibit nociceptive input.


Asunto(s)
Fibras Nerviosas Amielínicas/fisiología , Nocicepción/efectos de los fármacos , Oxitocina/farmacología , Células del Asta Posterior/fisiología , Receptores de Oxitocina/metabolismo , Médula Espinal/citología , Potenciales de Acción/efectos de los fármacos , Animales , Péptido Relacionado con Gen de Calcitonina/metabolismo , Canfanos/farmacología , Relación Dosis-Respuesta a Droga , Estimulación Eléctrica/efectos adversos , Formaldehído/toxicidad , Lectinas/metabolismo , Masculino , Nocicepción/fisiología , Dimensión del Dolor , Piperazinas/farmacología , Células del Asta Posterior/efectos de los fármacos , Desempeño Psicomotor/fisiología , Ratas , Ratas Wistar , Receptores de Oxitocina/análisis , Receptores de Oxitocina/antagonistas & inhibidores , Factores de Tiempo
19.
Brain Struct Funct ; 222(9): 3945-3958, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28528380

RESUMEN

The descending corticospinal (CS) projection has been considered a key element for motor control, which results from direct and indirect modulation of spinal cord pre-motor interneurons in the intermediate gray matter of the spinal cord, which, in turn, influences motoneurons in the ventral horn. The CS tract (CST) is also involved in a selective and complex modulation of sensory information in the dorsal horn. However, little is known about the spinal network engaged by the CST and the organization of CS projections that may encode different cortical outputs to the spinal cord. This study addresses the issue of whether the CS system exerts parallel control on different spinal networks, which together participate in sensorimotor integration. Here, we show that in the adult rat, two different and partially intermingled CS neurons in the sensorimotor cortex activate, with different time latencies, distinct spinal cord neurons located in the dorsal horn and intermediate zone of the same segment. The fact that different populations of CS neurons project in a segregated manner suggests that CST is composed of subsystems controlling different spinal cord circuits that modulate motor outputs and sensory inputs in a coordinated manner.


Asunto(s)
Corteza Cerebral/fisiología , Potenciales Evocados/fisiología , Tractos Piramidales/anatomía & histología , Tractos Piramidales/fisiología , Médula Espinal/fisiología , Potenciales de Acción/fisiología , Animales , Mapeo Encefálico , Corteza Cerebral/citología , Estimulación Eléctrica , Masculino , Neuronas/fisiología , Ratas , Ratas Wistar , Tiempo de Reacción , Médula Espinal/citología
20.
Neuropeptides ; 60: 51-60, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27449278

RESUMEN

The role of oxytocin (OXT) in pain modulation has been suggested. Indeed, hypothalamic paraventricular nuclei (PVN) electrical stimuli reduce the nociceptive neuronal activity (i.e., neuronal discharge associated with activation of Aδ- and C-fibers) of the spinal dorsal horn wide dynamic range (WDR) cells and nociceptive behavior. Furthermore, raphe magnus nuclei lesion reduces the PVN-induced antinociception, suggesting a functional interaction between the OXT and the serotoninergic system. The present study investigated in Wistar rats the potential role of spinal serotonergic mechanisms in the OXT- and PVN-induced antinociception. In long-term secondary mechanical allodynia and hyperalgesia induced by formalin or extracellular unitary recordings of the WDR cells we evaluated the role of 5-hydroxytryptamine (5-HT) effect on the OXT-induced antinociception. All drugs were given intrathecally (i.t.). OXT (1×10-5-1×10-4nmol) or 5-HT (1×10-3-1×10-1nmol) prevented the formalin-induced sensitization, an effect mimicked by PVN stimulation. Moreover, administration of OXT (1×10-5nmol) plus 5-HT (1×10-3nmol) at ineffective doses, produced antinociception. This effect was antagonized by: (i) d(CH2)5[Tyr(Me)2,Thr4,Tyr-NH29]OVT (oxytocin receptor antagonist; 2×10-2nmol); or (ii) methiothepin (a non-specific 5-HT1/2/5/6/7 receptor antagonist; 80nmol). Similar results were obtained with PVN stimulation plus 5-HT (5×10-5nmol). In WDR cell recordings, the PVN-induced antinociception was enhanced by i.t. 5-HT and partly blocked when the spinal cord was pre-treated with methiothepin (80nmol). Taken together, these results suggest that serotonergic mechanisms at the spinal cord level are partly involved in the OXT-induced antinociception.


Asunto(s)
Hiperalgesia/metabolismo , Nocicepción/fisiología , Oxitocina/farmacología , Dolor/metabolismo , Serotonina/farmacología , Médula Espinal/metabolismo , Animales , Estimulación Eléctrica , Formaldehído , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Masculino , Metiotepina/farmacología , Nocicepción/efectos de los fármacos , Oxitocina/uso terapéutico , Dolor/tratamiento farmacológico , Dimensión del Dolor , Ratas , Ratas Wistar , Receptores de Oxitocina/antagonistas & inhibidores , Antagonistas de la Serotonina/farmacología , Médula Espinal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...