Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Br J Pharmacol ; 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38715413

RESUMO

BACKGROUND AND PURPOSE: The voltage-gated sodium channel isoform NaV1.7 is a high-interest target for the development of non-opioid analgesics due to its preferential expression in pain-sensing neurons. NaV1.7 is also expressed in autonomic neurons, yet its contribution to involuntary visceral reflexes has received limited attention. The small molecule inhibitor ST-2560 was advanced into pain behaviour and cardiovascular models to understand the pharmacodynamic effects of selective inhibition of NaV1.7. EXPERIMENTAL APPROACH: Potency of ST-2560 at NaV1.7 and off-target ion channels was evaluated by whole-cell patch-clamp electrophysiology. Effects on nocifensive reflexes were assessed in non-human primate (NHP) behavioural models, employing the chemical capsaicin and mechanical stimuli. Cardiovascular parameters were monitored continuously in freely-moving, telemetered NHPs following administration of vehicle and ST-2560. KEY RESULTS: ST-2560 is a potent inhibitor (IC50 = 39 nM) of NaV1.7 in primates with ≥1000-fold selectivity over other isoforms of the human NaV1.x family. Following systemic administration, ST-2560 (0.1-0.3 mg·kg-1, s.c.) suppressed noxious mechanical- and chemical-evoked reflexes at free plasma concentrations threefold to fivefold above NaV1.7 IC50. ST-2560 (0.1-1.0 mg·kg-1, s.c.) also produced changes in haemodynamic parameters, most notably a 10- to 20-mmHg reduction in systolic and diastolic arterial blood pressure, at similar exposures. CONCLUSIONS AND IMPLICATIONS: Acute pharmacological inhibition of NaV1.7 is antinociceptive, but also has the potential to impact the cardiovascular system. Further work is merited to understand the role of NaV1.7 in autonomic ganglia involved in the control of heart rate and blood pressure, and the effect of selective NaV1.7 inhibition on cardiovascular function.

2.
J Pain ; 24(10): 1859-1874, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37271350

RESUMO

Traumatic brain injury (TBI) can cause acute and chronic pain along with motor, cognitive, and emotional problems. Although the mechanisms are poorly understood, previous studies suggest disruptions in endogenous pain modulation may be involved. Voluntary exercise after a TBI has been shown to reduce some consequences of injury including cognitive impairment. We hypothesized, therefore, that voluntary exercise could augment endogenous pain control systems in a rodent model of TBI. For these studies, we used a closed-head impact procedure in male mice modeling mild TBI. We investigated the effect of voluntary exercise on TBI-induced hindpaw nociceptive sensitization, diffuse noxious inhibitory control failure, and periorbital sensitization after bright light stress, a model of post-traumatic headache. Furthermore, we investigated the effects of exercise on memory, circulating markers of brain injury, neuroinflammation, and spinal cord gene expression. We observed that exercise significantly reduced TBI-induced hindpaw allodynia and periorbital allodynia in the first week following TBI. We also showed that exercise improved the deficits associated with diffuse noxious inhibitory control and reduced bright light stress-induced allodynia up to 2 months after TBI. In addition, exercise preserved memory and reduced TBI-induced increases in spinal BDNF, CXCL1, CXCL2, and prodynorphin expression, all genes previously linked to TBI-induced nociceptive sensitization. Taken together, our observations suggest that voluntary exercise may reduce pain after TBI by reducing TBI-induced changes in nociceptive signaling and preserving endogenous pain control systems. PERSPECTIVE: This article evaluates the effects of exercise on pain-related behaviors in a preclinical model of traumatic brain injury (TBI). The findings show that exercise reduces nociceptive sensitization, loss of diffuse noxious inhibitory control, memory deficits, and spinal nociception-related gene expression after TBI. Exercise may reduce or prevent pain after TBI.


Assuntos
Concussão Encefálica , Lesões Encefálicas Traumáticas , Dor Crônica , Cefaleia Pós-Traumática , Camundongos , Masculino , Animais , Cefaleia Pós-Traumática/complicações , Hiperalgesia/etiologia , Hiperalgesia/terapia , Lesões Encefálicas Traumáticas/complicações , Dor Crônica/complicações
3.
Pharmaceutics ; 14(5)2022 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-35631690

RESUMO

BACKGROUND AND PURPOSE: The intranasal administration of oxytocin (OT) reduces migraine headaches through activation of the oxytocin receptor (OTR). Magnesium ion (Mg2+) concentration is critical to the activation of the OTR, and a low serum Mg2+ concentration is predictive of a migraine headache. We, therefore, examined the functional impact of Mg2+ concentration on OT-OTR binding efficacy using two complimentary bioassays. EXPERIMENTAL APPROACH: Current clamp recordings of rat trigeminal ganglia (TG) neurons measured the impact of Mg2+ on an OT-induced reduction in excitability. In addition, we assessed the impact of Mg2+ on intranasal OT-induced craniofacial analgesia in rats. KEY RESULTS: While OT alone dose-dependently hyperpolarized TG neurons, decreasing their excitability, the addition of 1.75 mM Mg2+ significantly enhanced this effect. Similarly, while the intranasal application of OT produced dose-dependent craniofacial analgesia, Mg2+ significantly enhanced these effects. CONCLUSIONS AND IMPLICATIONS: OT efficacy may be limited by low ambient Mg2+ levels. The addition of Mg2+ to OT formulations may improve its efficacy in reducing headache pain as well as for other OT-dependent processes.

4.
Pharmaceutics ; 13(7)2021 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-34371778

RESUMO

Pain, particularly chronic pain, remains one of the most debilitating and difficult-to-treat conditions in medicine. Chronic pain is difficult to treat, in part because it is associated with plastic changes in the peripheral and central nervous systems. Polypeptides are linear organic polymers that are highly selective molecules for neurotransmitter and other nervous system receptors sites, including those associated with pain and analgesia, and so have tremendous potential in pain therapeutics. However, delivery of polypeptides to the nervous system is largely limited due to rapid degradation within the peripheral circulation as well as the blood-brain barrier. One strategy that has been shown to be successful in nervous system deposition of polypeptides is intranasal (IN) delivery. In this narrative review, we discuss the delivery of polypeptides to the peripheral and central nervous systems following IN administration. We briefly discuss the mechanism of delivery via the nasal-cerebral pathway. We review recent studies that demonstrate that polypeptides such as oxytocin, delivered IN, not only reach key pain-modulating regions in the nervous system but, in doing so, evoke significant analgesic effects. IN administration of polypeptides has tremendous potential to provide a non-invasive, rapid and effective method of delivery to the nervous system for chronic pain treatment and management.

5.
Transl Psychiatry ; 11(1): 388, 2021 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-34247185

RESUMO

There is evidence of the therapeutic potential of intranasal oxytocin for the treatment of pain and various psychiatric disorders, however, there is scant evidence that oxytocin reaches the brain. We quantified the concentration and distribution pattern of [125I]-radiolabeled oxytocin in the brains and peripheral tissues of rats after intranasal delivery using gamma counting and autoradiography, respectively. Radiolabel was detected in high concentrations in the trigeminal and olfactory nerves as well as in brain regions along their trajectories. Considerable concentrations were observed in the blood, however, relatively low levels of radiolabel were measured in peripheral tissues. The addition of a mucoadhesive did not enhance brain concentrations. These results provide support for intranasal OT reaching the brain via the olfactory and trigeminal neural pathways. These findings will inform the design and interpretation of clinical studies with intranasal oxytocin.


Assuntos
Transtornos Mentais , Ocitocina , Administração Intranasal , Animais , Encéfalo , Dor , Ratos
6.
Headache ; 61(7): 1051-1059, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34125955

RESUMO

OBJECTIVE: To highlight the emerging understanding of oxytocin (OT) and oxytocin receptors (OTRs) in modulating menstrual-related migraine (MRM). BACKGROUND: MRM is highly debilitating and less responsive to therapy, and attacks are of longer duration than nonmenstrually related migraine. A clear understanding of the mechanisms underlying MRM is lacking. METHODS: We present a narrative literature review on the developing understanding of the role of OT and the OTR in MRM. Literature on MRM on PubMed/MEDLINE database including clinical trials and basic science publications was reviewed using specific keywords. RESULTS: OT is a cyclically released hypothalamic hormone/neurotransmitter that binds to the OTR resulting in inhibition of trigeminal neuronal excitability that can promote migraine pain including that of MRM. Estrogen regulates OT release as well as expression of the OTR. Coincident with menstruation, levels of both estrogen and OT decrease. Additionally, other serum biochemical factors, including magnesium and cholesterol, which positively modulate the affinity of OT for OTRs, both decrease during menstruation. Thus, during menstruation, multiple menstrually associated factors may lead to decreased circulating OT levels, decreased OT affinity for OTR, and decreased expression of the trigeminal OTR. Consistent with the view of migraine as a threshold disorder, these events may collectively result in decreased inhibition promoting lower thresholds for activation of meningeal trigeminal nociceptors and increasing the likelihood of an MRM attack. CONCLUSION: Trigeminal OTR may thus be a novel target for the development of MRM therapeutics.


Assuntos
Estrogênios/metabolismo , Ciclo Menstrual/metabolismo , Distúrbios Menstruais/metabolismo , Transtornos de Enxaqueca/metabolismo , Ocitocina/metabolismo , Receptores de Ocitocina/metabolismo , Feminino , Humanos
7.
Pain ; 162(4): 1250-1261, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33086288

RESUMO

ABSTRACT: The voltage-gated sodium channel Nav1.7 is highly expressed in nociceptive afferents and is critically involved in pain signal transmission. Nav1.7 is a genetically validated pain target in humans because loss-of-function mutations cause congenital insensitivity to pain and gain-of-function mutations cause severe pain syndromes. Consequently, pharmacological inhibition has been investigated as an analgesic therapeutic strategy. We describe a small molecule Nav1.7 inhibitor, ST-2530, that is an analog of the naturally occurring sodium channel blocker saxitoxin. When evaluated against human Nav1.7 by patch-clamp electrophysiology using a protocol that favors the resting state, the Kd of ST-2530 was 25 ± 7 nM. ST-2530 exhibited greater than 500-fold selectivity over human voltage-gated sodium channel isoforms Nav1.1-Nav1.6 and Nav1.8. Although ST-2530 had lower affinity against mouse Nav1.7 (Kd = 250 ± 40 nM), potency was sufficient to assess analgesic efficacy in mouse pain models. A 3-mg/kg dose administered subcutaneously was broadly analgesic in acute pain models using noxious thermal, mechanical, and chemical stimuli. ST-2530 also reversed thermal hypersensitivity after a surgical incision on the plantar surface of the hind paw. In the spared nerve injury model of neuropathic pain, ST-2530 transiently reversed mechanical allodynia. These analgesic effects were demonstrated at doses that did not affect locomotion, motor coordination, or olfaction. Collectively, results from this study indicate that pharmacological inhibition of Nav1.7 by a small molecule agent with affinity for the resting state of the channel is sufficient to produce analgesia in a range of preclinical pain models.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.7 , Saxitoxina , Analgésicos/farmacologia , Analgésicos/uso terapêutico , Animais , Camundongos , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Canal de Sódio Disparado por Voltagem NAV1.8/genética , Isoformas de Proteínas , Bloqueadores dos Canais de Sódio/farmacologia , Bloqueadores dos Canais de Sódio/uso terapêutico
8.
Cereb Cortex ; 30(6): 3644-3654, 2020 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-32108220

RESUMO

Hypnosis is the oldest form of Western psychotherapy and a powerful evidence-based treatment for numerous disorders. Hypnotizability is variable between individuals; however, it is a stable trait throughout adulthood, suggesting that neurophysiological factors may underlie hypnotic responsiveness. One brain region of particular interest in functional neuroimaging studies of hypnotizability is the anterior cingulate cortex (ACC). Here, we examined the relationships between the neurochemicals, GABA, and glutamate, in the ACC and hypnotizability in healthy individuals. Participants underwent a magnetic resonance imaging (MRI) session, whereby T1-weighted anatomical and MEGA-PRESS spectroscopy scans were acquired. Voxel placement over the ACC was guided by a quantitative meta-analysis of functional neuroimaging studies of hypnosis. Hypnotizability was assessed using the Hypnotic Induction Profile (HIP), and self-report questionnaires to assess absorption (TAS), dissociation (DES), and negative affect were completed. ACC GABA concentration was positively associated with HIP scores such that the higher the GABA concentration, the more hypnotizable an individual. An exploratory analysis of questionnaire subscales revealed a negative relationship between glutamate and the absorption and imaginative involvement subscale of the DES. These results provide a putative neurobiological basis for individual differences in hypnotizability and can inform our understanding of treatment response to this growing psychotherapeutic tool.


Assuntos
Ácido Glutâmico/metabolismo , Giro do Cíngulo/metabolismo , Hipnose , Individualidade , Ácido gama-Aminobutírico/metabolismo , Adulto , Feminino , Giro do Cíngulo/diagnóstico por imagem , Humanos , Imageamento por Ressonância Magnética , Espectroscopia de Ressonância Magnética , Masculino , Inquéritos e Questionários , Adulto Jovem
9.
J Pain Res ; 11: 1991-1998, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30288094

RESUMO

PURPOSE: Noninvasive cerebral optical spectrometry is a promising candidate technology for the objective assessment physiological changes during pain perception. This study's primary objective was to test if there was a significant correlation between the changes in physiological parameters as measured by a cerebral optical spectrometry-based algorithm (real-time objective pain assessment [ROPA]) and subjective pain ratings obtained from volunteers and laboring women. Secondary aims were performance assessment using linear regression and receiver operating curve (ROC) analysis. PATIENTS AND METHODS: Prospective cohort study performed in Human Pain Laboratory and Labor and Delivery Unit. After institutional review board approval, we evaluated ROPA in volunteers undergoing the cold pressor test and in laboring women before and after epidural or combined spinal epidural placement. Linear regression was performed to measure correlations. ROCs and corresponding areas under the ROCs (AUC), as well as Youden's indices, as a measure of diagnostic effectiveness, were calculated. RESULTS: Correlations between numeric rating scale or visual analog scale and ROPA were significant for both volunteers and laboring women. AUCs for both volunteers and laboring women with numeric rating scale and visual analog scale subjective pain ratings as ground truth revealed at least good (AUC: 70%-79%) to excellent (AUC >90%) distinction between clinically meaningful pain severity differentiations (no/mild-moderate-severe). CONCLUSION: Cerebral Optical Spectrometry-based ROPA significantly correlated with subjectively reported pain in volunteers and laboring women, and could be a useful monitor for clinical circumstances where direct assessment is not available, or to complement patient-reported pain scores.

10.
Headache ; 58(4): 545-558, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29266199

RESUMO

BACKGROUND: Approximately 1.7 million Americans sustain a traumatic brain injury (TBI) each year and chronic pain is a common complication. OBJECTIVE: We studied the effects of intranasally administered oxytocin as a potential treatment for chronic pain in an animal model of mild TBI. METHODS: The lateral fluid percussion model of mild TBI was chosen for this purpose and after exposure to mild TBI the rats (n = 12) developed hind paw and facial allodynia compared to sham animals (n = 6). Oxytocin or a vehicle was afterwards administered intranasally and reactive pain was assessed by hind paw and facial von Frey testing. Some animals received the oxytocin receptor antagonist, atosiban, in addition to oxytocin/vehicle treatment (n = 12). The effect of oxytocin on ongoing and spontaneous pain was examined through conditioned place preference testing. To determine whether the effects of intranasal oxytocin could be attributed to delivery via the peripheral blood stream, some TBI animals received an intravenous injection of the same oxytocin dose that was given intranasally. ELISA immunoassays were carried out (n = 6) to measure concentrations of oxytocin in the trigeminal ganglia, pons, spinal cord, and olfactory bulb after intranasal administration and evaluate the most likely route of entry. RESULTS: These studies confirmed that the fluid percussion model can be used to study post-TBI facial allodynia. Oxytocin attenuated both reactive and spontaneous, ongoing non-reactive pain following mild TBI for at least 3-4 hours after intranasal administration by binding to OT or VA1-receptors most likely by a peri-trigeminal nerve mediated uptake. CONCLUSIONS: Intranasal oxytocin attenuates measures of reactive and non-reactive pain in a model of mild TBI and may represent a novel treatment for chronic pain in TBI patients.


Assuntos
Comportamento Animal/efeitos dos fármacos , Concussão Encefálica/complicações , Dor Crônica/tratamento farmacológico , Dor Facial/tratamento farmacológico , Hiperalgesia/tratamento farmacológico , Ocitocina/farmacologia , Administração Intranasal , Administração Intravenosa , Animais , Dor Crônica/etiologia , Modelos Animais de Doenças , Dor Facial/etiologia , Antagonistas de Hormônios/farmacologia , Hiperalgesia/etiologia , Masculino , Ocitocina/administração & dosagem , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Receptores de Ocitocina/antagonistas & inibidores , Vasotocina/análogos & derivados , Vasotocina/farmacologia
11.
Pain ; 158(11): 2181-2188, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29028747

RESUMO

Lysophosphatidic acid (LPA) is a bioactive lipid that impacts neurological outcomes after neurotrauma by inhibiting neuroregeneration, promoting inflammation, and contributing to behavioral deficits. Blocking LPA signaling with a novel anti-LPA monoclonal antibody (mAb) is neuroprotective after traumatic brain injury (TBI) if given to injured animals whose blood-brain barrier (BBB) has been compromised. It is hypothesized that the anti-LPA mAb could improve chronic pain initiated by TBI. However, poor brain penetration after systemic application of the antibody makes access to the central nervous system (CNS) problematic in situations where the BBB is intact. Our experiments investigated whether intranasal delivery of the anti-LPA mAb could bypass the BBB, allowing for direct entry of the antibody to certain areas of the CNS. When the humanized anti-LPA mAb, LT3114, was intranasally applied to injured rats within 30 minutes after mild TBI using the central lateral percussion model, enzyme-linked immunospecific assay and immunohistochemistry demonstrated antibody uptake to several areas in the CNS, including the area of cortical injury, the corpus callosum, cerebellum, and the subventricular region. Compared with control rats that received LT3114 but no TBI, TBI rats demonstrated significantly higher concentrations of intranasally administered LT3114 antibody in some tissues. In behavioral studies, a significant attenuation of mechanical allodynia after TBI was observed in the anti-LPA treatment group (P = 0.0079), when compared with vehicle controls within 14 days after TBI. These results suggest that intranasal application of the anti-LPA antibody directly accesses CNS sites involved in TBI-related pain and that this access attenuates pain sequelae to the neurotrauma.


Assuntos
Anticorpos/administração & dosagem , Lesões Encefálicas Traumáticas/complicações , Hiperalgesia/etiologia , Hiperalgesia/terapia , Lisofosfolipídeos/imunologia , Administração Intranasal , Animais , Anticorpos/sangue , Anticorpos/líquido cefalorraquidiano , Encéfalo/metabolismo , Encéfalo/patologia , Lesões Encefálicas Traumáticas/sangue , Lesões Encefálicas Traumáticas/líquido cefalorraquidiano , Lesões Encefálicas Traumáticas/patologia , Modelos Animais de Doenças , Masculino , Medição da Dor , Ratos , Ratos Sprague-Dawley
12.
Toxicol Sci ; 159(1): 76-85, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28903493

RESUMO

The longevity of pain after surgery is debilitating and limits the recovery of patients. AYX1 is a double-stranded, unprotected, 23 base-pair oligonucleotide designed to reduce acute post-surgical pain and prevent its chronification with a single intrathecal perioperative dose. AYX1 mimics the DNA sequence normally bound by EGR1 on chromosomes, a transcription factor transiently induced in the dorsal root ganglia-spinal cord network following a noxious input. AYX1 binds to EGR1 and prevents it from launching waves of gene regulation that are necessary to maintain pain over time. A formulation suitable for an intrathecal injection of AYX1 was developed, including a specific ratio of AYX1 and calcium so the ionic homeostasis of the cerebrospinal fluid is maintained and no impact on neuromuscular control is produced upon injection. A GLP toxicology study in naïve Sprague Dawley rats was conducted using 3 dose levels up to the maximum feasible dose. Clinical observations, neurobehavioral observations, clinical pathology and histopathology of the nervous system and peripheral tissues were conducted. An additional nonGLP study was conducted in the spared nerve injury model of chronic neuropathic pain in which EGR1 is induced in the dorsal root ganglia and spinal cord. Similar testing was performed, including a modified Irwin test to assess a potential impact of AYX1 on autonomic nervous system responses, locomotion, activity, arousal, sensorimotor, and neuromuscular function. No AYX1-related adverse events were observed in any of the studies and the no-observed-adverse-effect-level was judged to be the maximum feasible dose.


Assuntos
DNA/administração & dosagem , DNA/toxicidade , Animais , Comportamento Animal/efeitos dos fármacos , Cálcio/metabolismo , Líquido Cefalorraquidiano , DNA/metabolismo , Relação Dose-Resposta a Droga , Composição de Medicamentos , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Feminino , Homeostase , Injeções Espinhais , Masculino , Neuralgia/prevenção & controle , Ratos , Ratos Sprague-Dawley
13.
Theranostics ; 7(11): 2794-2805, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28824716

RESUMO

The ability to locate nerve injury and ensuing neuroinflammation would have tremendous clinical value for improving both the diagnosis and subsequent management of patients suffering from pain, weakness, and other neurologic phenomena associated with peripheral nerve injury. Although several non-invasive techniques exist for assessing the clinical manifestations and morphological aspects of nerve injury, they often fail to provide accurate diagnoses due to limited specificity and/or sensitivity. Herein, we describe a new imaging strategy for visualizing a molecular biomarker of nerve injury/neuroinflammation, i.e., the sigma-1 receptor (S1R), in a rat model of nerve injury and neuropathic pain. The two-fold higher increase of S1Rs was shown in the injured compared to the uninjured nerve by Western blotting analyses. With our novel S1R-selective radioligand, [18F]FTC-146 (6-(3-[18F]fluoropropyl)-3-(2-(azepan-1-yl)ethyl)benzo[d]thiazol-2(3H)-one), and positron emission tomography-magnetic resonance imaging (PET/MRI), we could accurately locate the site of nerve injury created in the rat model. We verified the accuracy of this technique by ex vivo autoradiography and immunostaining, which demonstrated a strong correlation between accumulation of [18F]FTC-146 and S1R staining. Finally, pain relief could also be achieved by blocking S1Rs in the neuroma with local administration of non-radioactive [19F]FTC-146. In summary, [18F]FTC-146 S1R PET/MR imaging has the potential to impact how we diagnose, manage and treat patients with nerve injury, and thus warrants further investigation.


Assuntos
Imageamento por Ressonância Magnética/métodos , Neuralgia/diagnóstico por imagem , Neuralgia/patologia , Nervos Periféricos/diagnóstico por imagem , Nervos Periféricos/patologia , Tomografia por Emissão de Pósitrons/métodos , Receptores sigma/análise , Animais , Azepinas/administração & dosagem , Benzotiazóis/administração & dosagem , Modelos Animais de Doenças , Radioisótopos de Flúor/administração & dosagem , Marcação por Isótopo , Masculino , Neuroma/complicações , Ratos Sprague-Dawley , Receptor Sigma-1
14.
Mol Pain ; 13: 1744806917727917, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28814144

RESUMO

Background: Nociception is maintained by genome-wide regulation of transcription in the dorsal root ganglia­spinal cord network. Hence, transcription factors constitute a promising class of targets for breakthrough pharmacological interventions to treat chronic pain. DNA decoys are oligonucleotides and specific inhibitors of transcription factor activities. A methodological series of in vivo­in vitro screening cycles was performed with decoy/transcription factor couples to identify targets capable of producing a robust and long-lasting inhibition of established chronic pain. Decoys were injected intrathecally and their efficacy was tested in the spared nerve injury and chronic constriction injury models of chronic pain in rats using repetitive von Frey testing. Results: Results demonstrated that a one-time administration of decoys binding to the Kruppel-like transcription factors (KLFs) 6, 9, and 15 produces a significant and weeks­month long reduction in mechanical hypersensitivity compared to controls. In the spared nerve injury model, decoy efficacy was correlated to its capacity to bind KLF15 and KLF9 at a specific ratio, while in the chronic constriction injury model, efficacy was correlated to the combined binding capacity to KLF6 and KLF9. AYX2, an 18-bp DNA decoy binding KLF6, KLF9, and KLF15, was optimized for clinical development, and it demonstrated significant efficacy in these models. Conclusions: These data highlight KLF6, KLF9, and KLF15 as transcription factors required for the maintenance of chronic pain and illustrate the potential therapeutic benefits of AYX2 for the treatment of chronic pain.


Assuntos
Dor Crônica/tratamento farmacológico , Fator 6 Semelhante a Kruppel/efeitos dos fármacos , Fatores de Transcrição Kruppel-Like/efeitos dos fármacos , Animais , Dor Crônica/metabolismo , Modelos Animais de Doenças , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Oligonucleotídeos/metabolismo , Ratos Sprague-Dawley , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo
15.
Mol Pain ; 13: 1744806917730212, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28845733

RESUMO

Abstract: Chronic pain after traumatic brain injury (TBI) is very common, but the mechanisms linking TBI to pain and the pain-related interactions of TBI with peripheral injuries are poorly understood. Chemokine receptors play an important role in both pain and brain injury. In the current work, we pursued the hypothesis that the epigenetically regulated CXC chemokine receptor 2 (CXCR2) is a crucial modulator of nociceptive sensitization induced by TBI. For these studies, we used the rat lateral fluid percussion model of TBI. Histone actyltransferase activity was blocked using anacardic acid beginning immediately following injury, or delayed for seven days prior to administration. The selective CXCR2 antagonist SCH527123 administered systemically or intrathecally was used to probe the role of chemokine signaling on mechanical hindpaw sensitization after TBI. The expression of the CXCR2 receptor was accomplished using real-time PCR, immunohistochemistry, and Western blotting, while epigenetic regulation was assessed using chromatin immunoprecipitation assay. The spinal levels of several pain-related mediators including CXCL1, an endogenous ligand for CXCR2, as well as brain-derived neurotrophic factor and prodynorphin were measured by enzyme-linked immunosorbent assay. We observed that anacardic acid potently blocked and reversed mechanical hindpaw sensitization after TBI. The same drug was able to prevent the upregulation of CXCR2 after TBI, but did not affect the spinal expression of other pain mediators. On the other hand, both systemically and intrathecally administered SCH527123 reversed hindpaw allodynia after TBI. Most of the spinal CXCR2 appeared to be expressed by spinal cord neurons. Chromatin immunoprecipitation experiments demonstrated TBI-enhanced association of the CXCR2 promoter with acetylated-H3K9 histone protein that was also reversible using anacardic acid. Taken together, our findings suggested that TBI causes the upregulation of spinal CXCR2 through an epigenetic mechanism ultimately supporting nociceptive sensitization. The use of CXCR2 antagonists may, therefore, be useful in pain resulting from TBI.


Assuntos
Benzamidas/farmacologia , Lesões Encefálicas Traumáticas/metabolismo , Ciclobutanos/farmacologia , Hiperalgesia/metabolismo , Receptores de Interleucina-8B/metabolismo , Ácidos Anacárdicos/farmacologia , Animais , Lesões Encefálicas Traumáticas/complicações , Quimiocina CXCL1/metabolismo , Modelos Animais de Doenças , Hiperalgesia/tratamento farmacológico , Masculino , Ratos Sprague-Dawley , Receptores de Interleucina-8B/efeitos dos fármacos , Medula Espinal/metabolismo , Regulação para Cima
16.
Curr Opin Anaesthesiol ; 30(4): 458-465, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28590258

RESUMO

PURPOSE OF REVIEW: This article reviews the phenomenon of opioid-induced hyperalgesia (OIH) and its implications for clinical anesthesia. The goal of this review is to give an update on perioperative prevention and treatment strategies, based on findings in preclinical and clinical research. RECENT FINDINGS: Several systems have been suggested to be involved in the pathophysiology of OIH with a focus on the glutaminergic system. Very recently preclinical data revealed that peripheral µ-opioid receptors (MORs) are key players in the development of OIH and acute opioid tolerance (AOT). Peripheral MOR antagonists could, thus, become a new prevention/treatment option of OIH in the perioperative setting. Although the impact of OIH on postoperative pain seems to be moderate, recent evidence suggests that increased hyperalgesia following opioid treatment correlates with the risk of developing persistent pain after surgery. In clinical practice, distinction among OIH, AOT and acute opioid withdrawal remains difficult, especially because a specific quantitative sensory test to diagnose OIH has not been validated yet. SUMMARY: Since the immediate postoperative period is not ideal to initiate long-term treatment for OIH, the best strategy is to prevent its occurrence. A multimodal approach, including choice of opioid, dose limitations and addition of nonopioid analgesics, is recommended.


Assuntos
Analgésicos Opioides/efeitos adversos , Anestesia/efeitos adversos , Hiperalgesia/induzido quimicamente , Tolerância a Medicamentos , Humanos , Hiperalgesia/prevenção & controle , Hiperalgesia/terapia , Síndrome de Abstinência a Substâncias/fisiopatologia
17.
Neuroreport ; 28(11): 661-665, 2017 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-28562485

RESUMO

Postincisional hyperalgesia and allodynia play an important role in perioperative medicine. NaV1.7 sodium channel has proven to be a key player in several pain states, including acute, inflammatory, and neuropathic pain. This study investigated the effects of silencing NaV1.7 through Herpes-based gene therapy with an antisense transcript on pain states after incision of the skin in rodents. Seventy-six Balb/C mice were subdivided into six groups and were treated with no virus, control virus, or NaV1.7 antisense vector before lateral hindpaw skin incision or sham procedure. All mice were tested for mechanical allodynia, cold allodynia, and thermal hyperalgesia. For time series analysis, a two-way analysis of variance with post-hoc Bonferroni testing was used. After incision mice developed significant hypersensitivity to mechanical, cold, and heat stimuli. The NaV1.7 antisense vector blocked the hypersensitivity to mechanical, cold, and heat stimuli that was normally observed 24 and 48 h after incision. We demonstrated that a gene therapy-based NaV1.7 knockdown affects postincisional hyperalgesia and allodynia. The data provide evidence that the incision model leads to periwound hypersensitivity after incision and that application of the NaV1.7 antisense virus prevents this sensitization. This then, in turn, provides presumptive support to the hypothesis that overexpression of the NaV1.7 channel is an important mechanism underlying hyperalgesia and allodynia following skin incision.


Assuntos
Hiperalgesia/genética , Canal de Sódio Disparado por Voltagem NAV1.7/genética , Neuralgia/genética , Pele/lesões , Animais , Modelos Animais de Doenças , Gânglios Espinais/metabolismo , Hiperalgesia/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Canal de Sódio Disparado por Voltagem NAV1.7/metabolismo
18.
Headache ; 57 Suppl 2: 64-75, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28485846

RESUMO

This article reviews material presented at the 2016 Scottsdale Headache Symposium. This presentation provided scientific results and rationale for the use of intranasal oxytocin for the treatment of migraine headache. Results from preclinical experiments are reviewed, including in vitro experiments demonstrating that trigeminal ganglia neurons possess oxytocin receptors and are inhibited by oxytocin. Furthermore, most of these same neurons contain CGRP, the release of which is inhibited by oxytocin. Results are also presented which demonstrate that nasal oxytocin inhibits responses of trigeminal nucleus caudalis neurons to noxious stimulation using either noxious facial shock or nitroglycerin infusion. These studies led to testing the analgesic effect of intranasal oxytocin in episodic migraineurs-studies which did not meet their primary endpoint of pain relief at 2 h, but which were highly informative and led to additional rat studies wherein inflammation was found to dramatically upregulate the number of oxytocin receptors available on trigeminal neurons. This importance of inflammation was supported by a series of in vivo rat behavioral studies, which demonstrated a clear craniofacial analgesic effect when a pre-existing inflammatory injury was present. The significance of inflammation was further solidified by a small single-dose clinical study, which showed analgesic efficacy that was substantially stronger in chronic migraine patients that had not taken an anti-inflammatory drug within 24 h of oxytocin dosing. A follow-on open label study examining effects of one month of intranasal oxytocin dosing did show a reduction in pain, but a more impressive decrease in the frequency of headaches in both chronic and high frequency episodic migraineurs. This study led to a multicountry double blind, placebo controlled study studying whether, over 2 months of dosing, "as needed" dosing of intranasal oxytocin by chronic and high frequency migraineurs would reduce the frequency of their headaches compared to a 1-month baseline period. This study failed to meet its primary endpoint, due to an extraordinarily high placebo rate in the country of most of the patients (Chile), but was also highly informative, showing strong results in other countries and strong post hoc indications of efficacy. The results provide a strong argument for further development of intranasal oxytocin for migraine prophylaxis.


Assuntos
Transtornos de Enxaqueca/prevenção & controle , Ocitócicos/administração & dosagem , Ocitocina/administração & dosagem , Administração Intranasal , Animais , Humanos , Gânglio Trigeminal/efeitos dos fármacos
19.
Mol Pain ; 13: 1744806917703112, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28394696

RESUMO

Background AYX1 is an unmodified DNA-decoy designed to reduce acute post-surgical pain and its chronification with a single intrathecal dose at the time of surgery. AYX1 inhibits the transcription factor early growth response protein 1, which is transiently induced at the time of injury and triggers gene regulation in the dorsal root ganglia and spinal cord that leads to long-term sensitization and pain. This work characterizes the AYX1 dose-response profile in rats and the link to AYX1 pharmacokinetics and metabolism in the cerebrospinal fluid, dorsal root ganglia, and spinal cord. Results The effects of ascending dose-levels of AYX1 on mechanical hypersensitivity were measured in the spared nerve injury model of chronic pain and in a plantar incision model of acute post-surgical pain. AYX1 dose-response profile shows that efficacy rapidly increases from a minimum effective dose of ∼ 0.5 mg to a peak maximum effective dose of ∼ 1 mg. With further dose escalation, the efficacy paradoxically appears to decrease by ∼ 30% and then returns to full efficacy at the maximum feasible dose of ∼ 4 mg. The reduction of efficacy is associated to doses triggering a near-saturation of AYX1 metabolism by nucleases in the cerebrospinal fluid and a paradoxical reduction of AYX1 exposure during the period of early growth response protein 1 induction. This effect is overcome at higher doses that compensate for the effect of metabolism. Discussion AYX1 is a competitive antagonist of early growth response protein 1, which is consistent with the overall increased efficacy observed as dose-levels initially escalate. Chemically, AYX1 is unprotected against degradation by nucleases. The sensitivity to nucleases is reflected in a paradoxical reduction of efficacy in the dose-response curve. Conclusions These findings point to the importance of the nuclease environment of the cerebrospinal fluid to the research and development of AYX1 and other intrathecal nucleotide-based therapeutics.


Assuntos
Dor Aguda/prevenção & controle , Analgésicos , Dor Crônica/prevenção & controle , DNA , Oligonucleotídeos , Dor Aguda/etiologia , Analgésicos/administração & dosagem , Analgésicos/metabolismo , Analgésicos/farmacologia , Animais , Dor Crônica/etiologia , DNA/administração & dosagem , DNA/metabolismo , DNA/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Gânglios Espinais/efeitos dos fármacos , Oligonucleotídeos/metabolismo , Oligonucleotídeos/farmacologia , Oligonucleotídeos/uso terapêutico , Medição da Dor , Dor Pós-Operatória/complicações , Dor Pós-Operatória/prevenção & controle , Ratos , Ratos Sprague-Dawley , Medula Espinal/efeitos dos fármacos
20.
Pain ; 158(4): 649-659, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28072605

RESUMO

Oxytocin (OXT) is a neuropeptide hormone synthesized and secreted by hypothalamic neurons and has been reported to play a significant role in pain modulation. However, the mechanisms underlying OXT's antinociceptive effect on neuropathic pain are not fully understood. In this study, we examined the peripheral effect of OXT on mechanical hypersensitivity induced by partial ligation of the infraorbital nerve (PNL) in rats. Mechanical hypersensitivity in the whisker pad skin after PNL was attenuated by the direct administration of OXT into the trigeminal ganglion (TG). The proportion of vasopressin-1A receptor (V1A-R)-immunoreactive, but not OXT-receptor-immunoreactive, neurons significantly increased among TG neurons innervating the whisker pad skin after PNL. In a patch-clamp recording from TG neurons isolated from PNL rats, the resting membrane potential of OXT-treated neurons was significantly decreased, and the current thresholds of OXT-treated neurons for spike generation (rheobases) were significantly greater than those of vehicle-treated neurons. In addition, OXT increased voltage-gated K channel currents in PNL animals. Furthermore, intra-TG administration of a selective V1A-R antagonist reversed the OXT-induced alleviation of mechanical hypersensitivity, and coapplication of the antagonist opposed OXT's effects on the resting membrane potential, rheobase, and K current. These findings suggest that OXT is effective at suppressing TG neuronal hyperexcitability after nerve injury, likely by modulation of voltage-gated K channels through V1A-R. This signaling mechanism represents a potential therapeutic target for the treatment of orofacial neuropathic pain.


Assuntos
Dor Facial/complicações , Hiperalgesia , Ocitocina/uso terapêutico , Receptores de Vasopressinas/metabolismo , Gânglio Trigeminal/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Dor Facial/tratamento farmacológico , Antagonistas de Hormônios/farmacologia , Hiperalgesia/tratamento farmacológico , Hiperalgesia/etiologia , Hiperalgesia/patologia , Indóis/farmacologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Neurônios/efeitos dos fármacos , Limiar da Dor/efeitos dos fármacos , Pirrolidinas/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Ocitocina/metabolismo , Gânglio Trigeminal/patologia , Vibrissas/inervação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...