Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 183
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Pharmacol Toxicol ; 60: 257-274, 2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31914896

RESUMEN

Neuropathic pain (NeP) can result from sources as varied as nerve compression, channelopathies, autoimmune disease, and incision. By identifying the neurobiological changes that underlie the pain state, it will be clinically possible to exploit mechanism-based therapeutics for maximum analgesic effect as diagnostic accuracy is optimized. Obtaining sufficient knowledge regarding the neuroadaptive alterations that occur in a particular NeP state will result in improved patient analgesia and a mechanism-based, as opposed to a disease-based, therapeutic approach to facilitate target identification. This will rely on comprehensive disease pathology insight; our knowledge is vastly improving due to continued forward and back translational preclinical and clinical research efforts. Here we discuss the clinical aspects of neuropathy and currently used drugs whose mechanisms of action are outlined alongside their clinical use. Finally, we consider sensory phenotypes, patient clusters, and predicting the efficacy of an analgesic for neuropathy.


Asunto(s)
Analgésicos/farmacología , Neuralgia/tratamiento farmacológico , Animales , Humanos , Neuralgia/diagnóstico , Neuralgia/fisiopatología , Fenotipo , Investigación Biomédica Traslacional
2.
J Neurosci ; 41(44): 9141-9162, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34544832

RESUMEN

The potassium channel Kv1.6 has recently been implicated as a major modulatory channel subunit expressed in primary nociceptors. Furthermore, its expression at juxtaparanodes of myelinated primary afferents is induced following traumatic nerve injury as part of an endogenous mechanism to reduce hyperexcitability and pain-related hypersensitivity. In this study, we compared two mouse models of constitutive Kv1.6 knock-out (KO) achieved by different methods: traditional gene trap via homologous recombination and CRISPR-mediated excision. Both Kv1.6 KO mouse lines exhibited an unexpected reduction in sensitivity to noxious heat stimuli, to differing extents: the Kv1.6 mice produced via gene trap had a far more significant hyposensitivity. These mice (Kcna6lacZ ) expressed the bacterial reporter enzyme LacZ in place of Kv1.6 as a result of the gene trap mechanism, and we found that their central primary afferent presynaptic terminals developed a striking neurodegenerative phenotype involving accumulation of lipid species, development of "meganeurites," and impaired transmission to dorsal horn wide dynamic range neurons. The anatomic defects were absent in CRISPR-mediated Kv1.6 KO mice (Kcna6-/-) but were present in a third mouse model expressing exogenous LacZ in nociceptors under the control of a Nav1.8-promoted Cre recombinase. LacZ reporter enzymes are thus intrinsically neurotoxic to sensory neurons and may induce pathologic defects in transgenic mice, which has confounding implications for the interpretation of gene KOs using lacZ Nonetheless, in Kcna6-/- mice not affected by LacZ, we demonstrated a significant role for Kv1.6 regulating acute noxious thermal sensitivity, and both mechanical and thermal pain-related hypersensitivity after nerve injury.SIGNIFICANCE STATEMENT In recent decades, the expansion of technologies to experimentally manipulate the rodent genome has contributed significantly to the field of neuroscience. While introduction of enzymatic or fluorescent reporter proteins to label neuronal populations is now commonplace, often potential toxicity effects are not fully considered. We show a role of Kv1.6 in acute and neuropathic pain states through analysis of two mouse models lacking Kv1.6 potassium channels: one with additional expression of LacZ and one without. We show that LacZ reporter enzymes induce unintended defects in sensory neurons, with an impact on behavioral data outcomes. To summarize we highlight the importance of Kv1.6 in recovery of normal sensory function following nerve injury, and careful interpretation of data from LacZ reporter models.


Asunto(s)
Técnicas de Inactivación de Genes/efectos adversos , Genes Reporteros , Canal de Potasio Kv1.6/genética , Operón Lac , Neuralgia/metabolismo , Nociceptores/metabolismo , Animales , Sistemas CRISPR-Cas , Femenino , Técnicas de Inactivación de Genes/métodos , Integrasas/metabolismo , Canal de Potasio Kv1.6/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Nociceptores/patología , Sinapsis/metabolismo , Sinapsis/patología
3.
Eur J Neurosci ; 51(4): 952-962, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31518451

RESUMEN

Diffuse noxious inhibitory controls (DNIC) are a mechanism of endogenous descending pain modulation and are deficient in a large proportion of chronic pain patients. However, the pathways involved remain only partially determined with several cortical and brainstem structures implicated. This study examined the role of the dorsal reticular nucleus (DRt) and infralimbic (ILC) region of the medial prefrontal cortex in DNIC. In vivo electrophysiology was performed to record from dorsal horn lamina V/VI wide dynamic range neurones with left hind paw receptive fields in anaesthetised sham-operated and L5/L6 spinal nerve-ligated (SNL) rats. Evoked neuronal responses were quantified in the presence and absence of a conditioning stimulus (left ear clamp). In sham rats, DNIC were reproducibly recruited by a heterotopically applied conditioning stimulus, an effect that was absent in neuropathic rats. Intra-DRt naloxone had no effect on spinal neuronal responses to dynamic brush, punctate mechanical, evaporative cooling and heat stimuli in sham and SNL rats. In addition, intra-DRt naloxone blocked DNIC in sham rats, but had no effect in SNL rats. Intra-ILC lidocaine had no effect on spinal neuronal responses to dynamic brush, punctate mechanical, evaporative cooling and heat stimuli in sham and SNL rats. However, differential effects were observed in relation to the expression of DNIC; intra-ILC lidocaine blocked activation of DNIC in sham rats but restored DNIC in SNL rats. These data suggest that the ILC is not directly involved in mediating DNIC but can modulate its activation and that DRt involvement in DNIC requires opioidergic signalling.


Asunto(s)
Control Inhibidor Nocivo Difuso , Animales , Tronco Encefálico , Humanos , Ratas , Ratas Sprague-Dawley , Médula Espinal , Nervios Espinales
4.
J Neural Transm (Vienna) ; 127(4): 541-549, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31515656

RESUMEN

Descending controls link higher processing of noxious signals to modulation of spinal cord responses to their noxious inputs. It has become possible to study one key inhibitory system in animals and humans using one painful stimulus to attenuate another distant response and so eliciting diffuse noxious inhibitory controls (DNIC) or the human counterpart, conditioned pain modulation (CPM). Here, we discuss the neuronal pathways in both species, their pharmacology and examine changes in descending controls with a focus on osteoarthritis. We will also discuss the opposing descending facilitatory system. Strong parallels between DNIC and CPM emphasize the possibility of forward and reverse translation.


Asunto(s)
Encéfalo , Neurotransmisores/farmacología , Osteoartritis , Dolor , Médula Espinal , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/fisiopatología , Humanos , Vías Nerviosas/metabolismo , Vías Nerviosas/fisiopatología , Osteoartritis/tratamiento farmacológico , Osteoartritis/metabolismo , Osteoartritis/fisiopatología , Dolor/tratamiento farmacológico , Dolor/metabolismo , Dolor/fisiopatología , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo , Médula Espinal/fisiopatología
5.
Anesthesiology ; 132(4): 881-894, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31977518

RESUMEN

BACKGROUND: The anterior cingulate cortex and central nucleus of the amygdala connect widely with brainstem nuclei involved in descending modulation, including the rostral ventromedial medulla. Endogenous opioids in these circuits participate in pain modulation. The hypothesis was that a differential opioidergic role for the brain nuclei listed in regulation of spinal neuronal responses because separable effects on pain behaviors in awake animals were previously observed. METHODS: This study utilized in vivo electrophysiology to determine the effects of morphine microinjection into the anterior cingulate cortex, right or left central nucleus of the amygdala, or the rostral ventromedial medulla on spinal wide dynamic range neuronal responses in isoflurane-anesthetized, male Sprague-Dawley rats. Ongoing activity in the ventrobasal thalamus was also measured. In total, 33 spinal nerve ligated and 26 control age- and weight-matched control rats were used. RESULTS: Brainstem morphine reduced neuronal firing to 60-g von Frey stimulation in control rats (to 65 ± 12% of control response (means ± 95% CI), P < 0.001) with a greater inhibition in neuropathic rats (to 53 ± 17% of control response, P < 0.001). Contrasting anterior cingulate cortex morphine had only marginal modulatory effects on spinal neuronal responses with limited variance in effect between control and neuropathic rats. The inhibitory effects of morphine in the central nucleus of the amygdala were dependent on pain state and laterality; only right-side morphine reduced neuronal firing to 60-g stimulation in neuropathic rats (to 65 ± 14% of control response, P = 0.001). In addition, in neuropathic rats elevated ongoing neuronal activity in the ventral posterolateral thalamus was not inhibited by anterior cingulate cortex morphine, in contrast to evoked responses. CONCLUSIONS: Cumulatively the data support opioid modulation of evoked responses predominately through a lateralized output from the right amygdala, as well as from the brainstem that is enhanced in injured conditions. Minimal modulation of dorsal horn responses was observed after anterior cingulate cortex opioid administration regardless of injury state.


Asunto(s)
Amígdala del Cerebelo/efectos de los fármacos , Analgésicos Opioides/administración & dosificación , Morfina/administración & dosificación , Red Nerviosa/efectos de los fármacos , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Nervios Espinales/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Amígdala del Cerebelo/fisiología , Animales , Relación Dosis-Respuesta a Droga , Giro del Cíngulo/efectos de los fármacos , Giro del Cíngulo/fisiología , Masculino , Bulbo Raquídeo/efectos de los fármacos , Bulbo Raquídeo/fisiología , Microinyecciones/métodos , Red Nerviosa/fisiología , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Células del Asta Posterior/efectos de los fármacos , Células del Asta Posterior/fisiología , Ratas , Ratas Sprague-Dawley , Nervios Espinales/fisiología
6.
J Neurophysiol ; 116(1): 159-70, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27098028

RESUMEN

Neuropathic pain represents a substantial clinical challenge; understanding the underlying neural mechanisms and back-translation of therapeutics could aid targeting of treatments more effectively. The ventral posterior thalamus (VP) is the major termination site for the spinothalamic tract and relays nociceptive activity to the somatosensory cortex; however, under neuropathic conditions, it is unclear how hyperexcitability of spinal neurons converges onto thalamic relays. This study aimed to identify neural substrates of hypersensitivity and the influence of pregabalin on central processing. In vivo electrophysiology was performed to record from VP wide dynamic range (WDR) and nociceptive-specific (NS) neurons in anesthetized spinal nerve-ligated (SNL), sham-operated, and naive rats. In neuropathic rats, WDR neurons had elevated evoked responses to low- and high-intensity punctate mechanical stimuli, dynamic brushing, and innocuous and noxious cooling, but less so to heat stimulation, of the receptive field. NS neurons in SNL rats also displayed increased responses to noxious punctate mechanical stimulation, dynamic brushing, noxious cooling, and noxious heat. Additionally, WDR, but not NS, neurons in SNL rats exhibited substantially higher rates of spontaneous firing, which may correlate with ongoing pain. The ratio of WDR-to-NS neurons was comparable between SNL and naive/sham groups, suggesting relatively few NS neurons gain sensitivity to low-intensity stimuli leading to a "WDR phenotype." After neuropathy was induced, the proportion of cold-sensitive WDR and NS neurons increased, supporting the suggestion that changes in frequency-dependent firing and population coding underlie cold hypersensitivity. In SNL rats, pregabalin inhibited mechanical and heat responses but not cold-evoked or elevated spontaneous activity.


Asunto(s)
Analgésicos/farmacología , Neuralgia/fisiopatología , Neuronas/fisiología , Pregabalina/farmacología , Tálamo/fisiología , Potenciales de Acción/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Ligadura , Masculino , Microelectrodos , Neuralgia/tratamiento farmacológico , Neuronas/efectos de los fármacos , Estimulación Física , Ratas Sprague-Dawley , Nervios Espinales/efectos de los fármacos , Nervios Espinales/fisiopatología , Tálamo/efectos de los fármacos
7.
Eur J Neurosci ; 44(1): 1714-22, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27108664

RESUMEN

Increasing evidence suggests that botulinum neurotoxins (BoNTs) delivered into the skin and muscle in certain human and animal pain states may exert antinociceptive efficacy though their uptake and transport to central afferent terminals. Cleavage of soluble N-methylaleimide-sensitive attachment protein receptor by BoNTs can impede vesicular mediated neurotransmitter release as well as transport/insertion of channel/receptor subunits into plasma membranes, an effect that can reduce activity-evoked facilitation. Here, we explored the effects of intraplantar botulinum toxin- B (BoNT-B) on peripheral inflammation and spinal nociceptive processing in an inflammatory model of pain. C57BL/6 mice (male) received unilateral intraplantar BoNT (1 U, 30 µL) or saline prior to intraplantar carrageenan (20 µL, 2%) or intrathecal N-methyl-D-aspartate (NMDA), substance P or saline (5 µL). Intraplantar carrageenan resulted in edema and mechanical allodynia in the injected paw and increased phosphorylation of a glutamate subunit (pGluA1ser845) and a serine/threonine-specific protein kinase (pAktser473) in spinal dorsal horn along with an increased incidence of spinal c-Fos positive cells. Pre-treatment with intraplantar BoNT-B reduced carrageenan evoked: (i) allodynia, but not edema; (ii) pGluA1 and pAkt and (iii) c-Fos expression. Further, intrathecal NMDA and substance P each increased dorsal horn levels of pGluA1 and pAkt. Intraplantar BoNT-B inhibited NMDA, but not substance P evoked phosphorylation of GluA1 and Akt. These results suggest that intraplantar toxin is transported centrally to block spinal activation and prevent phosphorylation of a glutamate receptor subunit and a kinase, which otherwise contribute to facilitated states.


Asunto(s)
Analgésicos/farmacología , Toxinas Botulínicas Tipo A/farmacología , Nocicepción , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores AMPA/metabolismo , Asta Dorsal de la Médula Espinal/metabolismo , Analgésicos/administración & dosificación , Analgésicos/uso terapéutico , Animales , Toxinas Botulínicas Tipo A/administración & dosificación , Toxinas Botulínicas Tipo A/uso terapéutico , Carragenina/toxicidad , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/etiología , Hiperalgesia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , N-Metilaspartato/farmacología , Fosforilación , Proteínas Proto-Oncogénicas c-fos/metabolismo , Asta Dorsal de la Médula Espinal/fisiología , Sustancia P/farmacología
8.
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
9.
J Physiol ; 593(17): 4029-42, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26047369

RESUMEN

Translational research is key to bridging the gaps between preclinical findings and the patients, and a translational model of inflammatory pain will ideally induce both peripheral and central sensitisation, more effectively mimicking clinical pathophysiology in some chronic inflammatory conditions. We conducted a parallel investigation of two models of inflammatory pain, using ultraviolet B (UVB) irradiation alone and UVB irradiation with heat rekindling. We used rodent electrophysiology and human quantitative sensory testing to characterise nociceptive processing in the peripheral and central nervous systems in both models. In both species, UVB irradiation produces peripheral sensitisation measured as augmented evoked activity of rat dorsal horn neurones and increased perceptual responses of human subjects to mechanical and thermal stimuli. In both species, UVB with heat rekindling produces central sensitisation. UVB irradiation alone and UVB with heat rekindling are translational models of inflammation that produce peripheral and central sensitisation, respectively. The predictive value of laboratory models for human pain processing is crucial for improving translational research. The discrepancy between peripheral and central mechanisms of pain is an important consideration for drug targets, and here we describe two models of inflammatory pain that involve ultraviolet B (UVB) irradiation, which can employ peripheral and central sensitisation to produce mechanical and thermal hyperalgesia in rats and humans. We use electrophysiology in rats to measure the mechanically- and thermally-evoked activity of rat spinal neurones and quantitative sensory testing to assess human psychophysical responses to mechanical and thermal stimulation in a model of UVB irradiation and in a model of UVB irradiation with heat rekindling. Our results demonstrate peripheral sensitisation in both species driven by UVB irradiation, with a clear mechanical and thermal hypersensitivity of rat dorsal horn neurones and enhanced perceptual responses of human subjects to both mechanical and thermal stimulation. Additional heat rekindling produces markers of central sensitisation in both species, including enhanced receptive field sizes. Importantly, we also showed a correlation in the evoked activity of rat spinal neurones to human thermal pain thresholds. The parallel results in rats and humans validate the translational use of both models and the potential for such models for preclinical assessment of prospective analgesics in inflammatory pain states.


Asunto(s)
Quemaduras/psicología , Hiperalgesia/psicología , Percepción del Dolor , Dolor/psicología , Células del Asta Posterior/fisiología , Rayos Ultravioleta/efectos adversos , Adulto , Animales , Calor , Humanos , Hiperalgesia/fisiopatología , Inflamación/fisiopatología , Inflamación/psicología , Masculino , Dolor/fisiopatología , Umbral del Dolor , Estimulación Física , Psicofísica , Ratas Sprague-Dawley , Adulto Joven
10.
Eur J Neurosci ; 42(11): 3004-11, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26454262

RESUMEN

Cold hypersensitivity is evident in a range of neuropathies and can evoke sensations of paradoxical burning cold pain. Ciguatoxin poisoning is known to induce a pain syndrome caused by consumption of contaminated tropical fish that can persist for months and include pruritus and cold allodynia; at present no suitable treatment is available. This study examined, for the first time, the neural substrates and molecular components of Pacific ciguatoxin-2-induced cold hypersensitivity. Electrophysiological recordings of dorsal horn lamina V/VI wide dynamic range neurones were made in non-sentient rats. Subcutaneous injection of 10 nm ciguatoxin-2 into the receptive field increased neuronal responses to innocuous and noxious cooling. In addition, neuronal responses to low-threshold but not noxious punctate mechanical stimuli were also elevated. The resultant cold hypersensitivity was not reversed by 6-({2-[2-fluoro-6-(trifluoromethyl)phenoxy]-2-methylpropyl}carbamoyl)pyridine-3-carboxylic acid, an antagonist of transient receptor potential melastatin 8 (TRPM8). Both mechanical and cold hypersensitivity were completely prevented by co-injection with the Nav 1.8 antagonist A803467, whereas the transient receptor potential ankyrin 1 (TRPA1) antagonist A967079 only prevented hypersensitivity to innocuous cooling and partially prevented hypersensitivity to noxious cooling. In naive rats, neither innocuous nor noxious cold-evoked neuronal responses were inhibited by antagonists of Nav 1.8, TRPA1 or TRPM8 alone. Ciguatoxins may confer cold sensitivity to a subpopulation of cold-insensitive Nav 1.8/TRPA1-positive primary afferents, which could underlie the cold allodynia reported in ciguatera. These data expand the understanding of central spinal cold sensitivity under normal conditions and the role of these ion channels in this translational rat model of ciguatoxin-induced hypersensitivity.


Asunto(s)
Ciguatoxinas/toxicidad , Síndromes Periódicos Asociados a Criopirina/fisiopatología , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Neuronas/fisiología , Médula Espinal/fisiopatología , Canales Catiónicos TRPC/metabolismo , Canales Catiónicos TRPM/metabolismo , Compuestos de Anilina/farmacología , Animales , Frío , Síndromes Periódicos Asociados a Criopirina/tratamiento farmacológico , Modelos Animales de Enfermedad , Furanos/farmacología , Masculino , Microelectrodos , Neuronas/efectos de los fármacos , Neurotransmisores/farmacología , Ácidos Nicotínicos/farmacología , Oximas/farmacología , Ratas Sprague-Dawley , Médula Espinal/efectos de los fármacos , Canal Catiónico TRPA1 , Canales Catiónicos TRPC/antagonistas & inhibidores , Canales Catiónicos TRPM/antagonistas & inhibidores , Tacto , Bloqueadores del Canal de Sodio Activado por Voltaje/farmacología
11.
Pharmacol Rev ; 64(4): 939-71, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23023032

RESUMEN

A large number of pharmacological studies have used capsaicin as a tool to activate many physiological systems, with an emphasis on pain research but also including functions such as the cardiovascular system, the respiratory system, and the urinary tract. Understanding the actions of capsaicin led to the discovery its receptor, transient receptor potential (TRP) vanilloid subfamily member 1 (TRPV1), part of the superfamily of TRP receptors, sensing external events. This receptor is found on key fine sensory afferents, and so the use of capsaicin to selectively activate pain afferents has been exploited in animal studies, human psychophysics, and imaging studies. Its effects depend on the dose and route of administration and may include sensitization, desensitization, withdrawal of afferent nerve terminals, or even overt death of afferent fibers. The ability of capsaicin to generate central hypersensitivity has been valuable in understanding the consequences and mechanisms behind enhanced central processing of pain. In addition, capsaicin has been used as a therapeutic agent when applied topically, and antagonists of the TRPV1 receptor have been developed. Overall, the numerous uses for capsaicin are clear; hence, the rationale of this review is to bring together and discuss the different types of studies that exploit these actions to shed light upon capsaicin working both as a tool to understand pain but also as a treatment for chronic pain. This review will discuss the various actions of capsaicin and how it lends itself to these different purposes.


Asunto(s)
Capsaicina/administración & dosificación , Dolor/fisiopatología , Animales , Capsaicina/farmacocinética , Tracto Gastrointestinal/metabolismo , Humanos , Modelos Biológicos , Dolor/inducido químicamente , Dolor/tratamiento farmacológico , Sistema Respiratorio/metabolismo , Piel/metabolismo , Canales de Potencial de Receptor Transitorio/fisiología
12.
J Neurosci ; 33(42): 16412-26, 2013 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-24133248

RESUMEN

The α2δ-1 subunit of voltage-gated calcium channels is upregulated after sensory nerve injury and is also the therapeutic target of gabapentinoid drugs. It is therefore likely to play a key role in the development of neuropathic pain. In this study, we have examined mice in which α2δ-1 gene expression is disrupted, to determine whether α2δ-1 is involved in various modalities of nociception, and for the development of behavioral hypersensitivity after partial sciatic nerve ligation (PSNL). We find that naive α2δ-1(-/-) mice show a marked behavioral deficit in mechanical and cold sensitivity, but no change in thermal nociception threshold. The lower mechanical sensitivity is mirrored by a reduced in vivo electrophysiological response of dorsal horn wide dynamic range neurons. The CaV2.2 level is reduced in brain and spinal cord synaptosomes from α2δ-1(-/-) mice, and α2δ-1(-/-) DRG neurons exhibit lower calcium channel current density. Furthermore, a significantly smaller number of DRG neurons respond to the TRPM8 agonist menthol. After PSNL, α2δ-1(-/-) mice show delayed mechanical hypersensitivity, which only develops at 11 d after surgery, whereas in wild-type littermates it is maximal at the earliest time point measured (3 d). There is no compensatory upregulation of α2δ-2 or α2δ-3 after PSNL in α2δ-1(-/-) mice, and other transcripts, including neuropeptide Y and activating transcription factor-3, are upregulated normally. Furthermore, the ability of pregabalin to alleviate mechanical hypersensitivity is lost in PSNL α2δ-1(-/-) mice. Thus, α2δ-1 is essential for rapid development of mechanical hypersensitivity in a nerve injury model of neuropathic pain.


Asunto(s)
Canales de Calcio/genética , Hiperalgesia/genética , Neuralgia/genética , Traumatismos de los Nervios Periféricos/genética , Células Receptoras Sensoriales/fisiología , Animales , Canales de Calcio/metabolismo , Frío , Ganglios Espinales/metabolismo , Ganglios Espinales/fisiopatología , Hiperalgesia/metabolismo , Hiperalgesia/fisiopatología , Ratones , Ratones Noqueados , Neuralgia/metabolismo , Neuralgia/fisiopatología , Umbral del Dolor/fisiología , Traumatismos de los Nervios Periféricos/metabolismo , Traumatismos de los Nervios Periféricos/fisiopatología , Estimulación Física , Nervio Ciático/lesiones , Nervio Ciático/metabolismo , Nervio Ciático/fisiopatología
13.
J Pharmacol Exp Ther ; 349(1): 47-55, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24472724

RESUMEN

Abnormal cold sensitivity is a common feature of a range of neuropathies. In the murine somatosensory system, multiple aspects of cold sensitivity are dependent on TRPM8, both short term and in response to peripheral nerve injury. The specialized nature of cold-sensitive afferents and the restricted expression of TRPM8 render it an attractive target for the treatment of cold hypersensitivity. This current study examines the effect of a novel TRPM8 antagonist (M8-An) in naive and spinal nerve-ligated rats through behavioral and in vivo electrophysiological approaches. In vitro, M8-An inhibited icilin-evoked Ca(2+) currents in HEK293 cells stably expressing human TRPM8 with an IC(50) of 10.9 nM. In vivo, systemic M8-An transiently decreased core body temperature. Deep dorsal horn recordings were made in vivo from neurons innervating the hind paw. M8-An inhibited neuronal responses to innocuous and noxious cooling of the receptive field in spinal nerve-ligated rats but not in naive rats. No effect on neuronal responses to mechanical and heat stimulation was observed. In addition, M8-An also attenuated behavioral responses to cold but not mechanical stimulation after nerve ligation without affecting the uninjured contralateral response. The data presented here support a contribution of TRPM8 to the pathophysiology of cold hypersensitivity in this model and highlight the potential of the pharmacological block of TRPM8 in alleviating the associated symptoms.


Asunto(s)
Síndromes Periódicos Asociados a Criopirina/prevención & control , Ácidos Nicotínicos/uso terapéutico , Traumatismos de los Nervios Periféricos/complicaciones , Canales Catiónicos TRPM/antagonistas & inhibidores , Animales , Conducta Animal/efectos de los fármacos , Calcio/metabolismo , Síndromes Periódicos Asociados a Criopirina/etiología , Síndromes Periódicos Asociados a Criopirina/metabolismo , Modelos Animales de Enfermedad , Fenómenos Electrofisiológicos , Células HEK293 , Humanos , Masculino , Ácidos Nicotínicos/administración & dosificación , Ácidos Nicotínicos/farmacología , Traumatismos de los Nervios Periféricos/metabolismo , Traumatismos de los Nervios Periféricos/psicología , Ratas , Ratas Sprague-Dawley
14.
Ann Neurol ; 74(5): 630-6, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24018757

RESUMEN

Many pain states begin with damage to tissue and/or nerves in the periphery, leading to enhanced transmitter release within the spinal cord and central sensitization. Manifestations of this central sensitization are windup and long-term potentiation. Hyperexcitable spinal neurons show reduced thresholds, greater evoked responses, increased receptive field sizes, and ongoing stimulus-independent activity; these changes probably underlie the allodynia, hyperalgesia, and spontaneous pain seen in patients. Central sensitization is maintained by continuing input from the periphery, but also modulated by descending controls, both inhibitory and facilitatory, from the midbrain and brainstem. The projections of sensitized spinal neurons to the brain, in turn, alter the processing of painful messages by higher centers. Several mechanisms contribute to central sensitization. Repetitive activation of primary afferent C fibers leads to a synaptic strengthening of nociceptive transmission. It may also induce facilitation of non-nociceptive Aß fibers and nociceptive Aδ fibers, giving rise to dynamic mechanical allodynia and mechanical hyperalgesia. In postherpetic neuralgia and complex regional pain syndrome, for example, these symptoms are maintained and modulated by peripheral nociceptive input. Diagnosing central sensitization can be particularly difficult. In addition to the medical history, quantitative sensory testing and functional magnetic resonance imaging may be useful, but diagnostic criteria that include both subjective and objective measures of central augmentation are needed. Mounting evidence indicates that treatment strategies that desensitize the peripheral and central nervous systems are required. These should generally involve a multimodal approach, so that therapies may target the peripheral drivers of central sensitization and/or the central consequences.


Asunto(s)
Encéfalo/fisiopatología , Sensibilización del Sistema Nervioso Central/fisiología , Hiperalgesia/fisiopatología , Células Receptoras Sensoriales/fisiología , Médula Espinal/fisiopatología , Animales , Humanos , Hiperalgesia/diagnóstico
15.
Expert Opin Emerg Drugs ; 19(3): 329-41, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24793304

RESUMEN

INTRODUCTION: Neuropathic pain is a costly and disabling condition, which affects up to 8% of the population. Available therapies often provide incomplete pain relief and treatment-related side effects are common. Preclinical neuropathic pain models have facilitated identification of several promising targets, which have progressed to human clinical phases of evaluation. AREAS COVERED: A systematic database search yielded 25 new molecular entities with specified pharmacological mechanisms that have reached Phase II or III clinical trials. These include calcium channel antagonists, vanilloid receptor antagonists, potassium channel agonists, NMDA antagonists, novel opioid receptor agonists, histamine H3 receptor antagonists, a novel sodium channel antagonist, serotonin modulators, a novel acetylcholine receptor agonist, α-2b adrenoreceptor agonist, cannabinoid CB2 receptor agonist, nitric oxide synthase inhibitor, orexin receptor antagonist, angiotensin II 2 antagonist, imidazoline I2 receptor agonist, apoptosis inhibitor and fatty acid amide hydrolase inhibitor. EXPERT OPINION: Although the diversity of pharmacological mechanisms of interest emphasise the complexity of neuropathic pain transmission, the considerable number of agents under development reflect a continued enthusiasm in drug development for neuropathic pain. Ongoing enhancements in methodology of both preclinical and clinical research and closer translation in both directions are expected to more efficiently identify new agents, which will improve the management of neuropathic pain.


Asunto(s)
Analgésicos/uso terapéutico , Diseño de Fármacos , Neuralgia/tratamiento farmacológico , Analgésicos/efectos adversos , Analgésicos/farmacología , Animales , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Humanos , Neuralgia/fisiopatología
16.
Brain ; 141(1): 5-6, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29325048
17.
Pain ; 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38198231

RESUMEN

ABSTRACT: In humans and animals, high-frequency electrocutaneous stimulation (HFS) induces an "early long-term potentiation-like" sensitisation, where synaptic plasticity is underpinned by an ill-defined interaction between peripheral input and central modulatory processes. The relative contributions of these processes to the initial pain or nociceptive response likely differ from those that underpin development of the heightened response. To investigate the impact of HFS-induced hyperalgesia on pain and nociception in perception and neural terms, respectively, and to explore the impact of descending inhibitory pathway activation on the development of HFS-induced hyperalgesia, we performed parallel studies utilising identical stimuli to apply HFS concurrent to (1) a conditioned pain modulation paradigm during psychophysical testing in healthy humans or (2) a diffuse noxious inhibitory controls paradigm during in vivo electrophysiological recording of spinal neurones in healthy anaesthetised rats. High-frequency electrocutaneous stimulation alone induced enhanced perceptual responses to pinprick stimuli in cutaneous areas secondary to the area of electrical stimulation in humans and increased the excitability of spinal neurones which exhibited stimulus intensity-dependent coded responses to pinprick stimulation in a manner that tracked with human psychophysics, supporting their translational validity. Application of a distant noxious conditioning stimulus during HFS did not alter perceived primary or secondary hyperalgesia in humans or the development of primary or secondary neuronal hyperexcitability in rats compared with HFS alone, suggesting that, upon HFS-response initiation in a healthy nervous system, excitatory signalling escapes inhibitory control. Therefore, in this model, dampening facilitatory mechanisms rather than augmenting top-down inhibitions could prevent pain development.

18.
J Pharmacol Exp Ther ; 344(3): 696-707, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23297162

RESUMEN

Osteoarthritis (OA) is a chronic joint disorder whose principal symptom is chronic pain. Current analgesics are inadequate and the mechanisms contributing to this pain are poorly understood but likely to include both local joint changes and central consequences. These studies used monoamine receptor agents combined with behavioral studies and single-unit dorsal horn recordings to examine whether descending noradrenergic and serotonergic inhibitions are altered in the monosodium iodoacetate model of OA pain, and whether increasing these inhibitions with the serotonin/noradrenaline reuptake inhibitor milnacipran can attenuate the attendant hypersensitivity. Early and late in the course of this model, milnacipran (s.c.) reduced behavioral hypersensitivity, and inhibited evoked responses from sensitized dorsal horn neurons. In naïve animals and the early, but not late, phase of the model, spinal administration of the α(2)-adrenoceptor antagonist atipamezole fully reversed this neuronal inhibition, whereas atipamezole administered alone revealed that endogenous noradrenergic inhibition was reduced in the late phase. Blocking spinal 5-hydroxytryptamine-7 receptors with (2R)-1-[(3-hydroxyphenyl)sulfonyl]-2-[2-(4-methyl-1-piperidinyl)ethyl]pyrrolidine hydrochloride suggested that the effects of milnacipran in the late phase were partly mediated by these receptors, and that descending serotonergic inhibition was increased in this phase. An opioidergic mechanism behind the effects of milnacipran was indicated by a partial reversal of these effects with naloxone. These studies demonstrate antinociceptive effects for milnacipran in a model of OA pain, whose effects come via descending serotonergic and noradrenergic, as well as opioidergic, pathways. Variations in the activity of these pathways over the course of this model may contribute to the presence of behavioral hypersensitivity and determine through which endogenous systems milnacipran exerts its effects.


Asunto(s)
Analgésicos/farmacología , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/etiología , Ciclopropanos/farmacología , Osteoartritis/complicaciones , Neuronas Adrenérgicas/efectos de los fármacos , Neuronas Adrenérgicas/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 2/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Dolor Crónico/metabolismo , Imidazoles/farmacología , Masculino , Milnaciprán , Naloxona/farmacología , Antagonistas de Narcóticos , Norepinefrina/antagonistas & inhibidores , Norepinefrina/metabolismo , Osteoartritis/inducido químicamente , Osteoartritis/metabolismo , Células del Asta Posterior/efectos de los fármacos , Células del Asta Posterior/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Opioides/metabolismo , Receptores de Serotonina/metabolismo , Neuronas Serotoninérgicas/efectos de los fármacos , Neuronas Serotoninérgicas/metabolismo , Serotonina/metabolismo , Antagonistas de la Serotonina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Médula Espinal/efectos de los fármacos , Médula Espinal/metabolismo
19.
Pain ; 164(6): 1264-1279, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36524581

RESUMEN

ABSTRACT: Neuronal N-type (Ca V 2.2) voltage-gated calcium channels are essential for neurotransmission from primary afferent terminals in the dorsal horn. In this study, we have used a knockin mouse containing Ca V 2.2 with an inserted extracellular hemagglutinin tag (Ca V 2.2_HA), to visualise the pattern of expression of endogenous Ca V 2.2 in dorsal root ganglion (DRG) neurons and their primary afferents in the dorsal horn. We examined the effect of partial sciatic nerve ligation (PSNL) and found an increase in Ca V 2.2_HA only in large and medium dorsal root ganglion neurons and also in deep dorsal horn synaptic terminals. Furthermore, there is a parallel increase in coexpression with GFRα1, present in a population of low threshold mechanoreceptors, both in large DRG neurons and in their terminals. The increased expression of Ca V 2.2_HA in these DRG neurons and their terminals is dependent on the presence of the auxiliary subunit α 2 δ-1, which is required for channel trafficking to the cell surface and to synaptic terminals, and it likely contributes to enhanced synaptic transmission at these synapses following PSNL. By contrast, the increase in GFRα1 is not altered in α 2 δ-1-knockout mice. We also found that following PSNL, there is patchy loss of glomerular synapses immunoreactive for Ca V 2.2_HA and CGRP or IB4, restricted to the superficial layers of the dorsal horn. This reduction is not dependent on α 2 δ-1 and likely reflects partial deafferentation of C-nociceptor presynaptic terminals. Therefore, in this pain model, we can distinguish 2 different events affecting specific DRG terminals, with opposite consequences for Ca V 2.2_HA expression and function in the dorsal horn.


Asunto(s)
Ganglios Espinales , Mecanorreceptores , Enfermedades del Sistema Nervioso Periférico , Animales , Ratones , Ratas , Ganglios Espinales/metabolismo , Nociceptores/metabolismo , Enfermedades del Sistema Nervioso Periférico/metabolismo , Ratas Sprague-Dawley
20.
Pain ; 164(1): 3-13, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-35580314

RESUMEN

ABSTRACT: Ambroxol is a multifaceted drug with primarily mucoactive and secretolytic actions, along with anti-inflammatory, antioxidant, and local anaesthetic properties. It has a long history of use in the treatment of respiratory tract diseases and has shown to be efficacious in relieving sore throat. In more recent years, ambroxol has gained interest for its potential usefulness in treating neuropathic pain. Research into this area has been slow, despite clear preclinical evidence to support its primary analgesic mechanism of action-blockade of voltage-gated sodium (Na v ) channels in sensory neurons. Ambroxol is a commercially available inhibitor of Na v 1.8, a crucial player in the pathophysiology of neuropathic pain, and Na v 1.7, a particularly exciting target for the treatment of chronic pain. In this review, we discuss the analgesic mechanisms of action of ambroxol, as well as proposed synergistic properties, followed by the preclinical and clinical results of its use in the treatment of persistent pain and neuropathic pain symptoms, including trigeminal neuralgia, fibromyalgia, and complex regional pain syndrome. With its well-established safety profile, extensive preclinical and clinical drug data, and early evidence of clinical effectiveness, ambroxol is an old drug worthy of further investigation for repurposing. As a patent-expired drug, a push is needed to progress the drug to clinical trials for neuropathic pain. We encourage the pharmaceutical industry to look at patented drug formulations and take an active role in bringing an optimized version for neuropathic pain to market.


Asunto(s)
Ambroxol , Neuralgia , Humanos , Ambroxol/uso terapéutico , Ambroxol/farmacología , Neuralgia/tratamiento farmacológico , Analgésicos/uso terapéutico , Anestésicos Locales/uso terapéutico , Dimensión del Dolor
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA