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













Base de datos
Intervalo de año de publicación
1.
Neuroscience ; 541: 64-76, 2024 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-38307407

RESUMEN

Corticosteroids are commonly used in the treatment of inflammatory low back pain, and their nominal target is the glucocorticoid receptor (GR) to relieve inflammation. They can also have similar potency at the mineralocorticoid receptor (MR). The MR has been shown to be widespread in rodent and human dorsal root ganglia (DRG) neurons and non-neuronal cells, and when MR antagonists are administered during a variety of inflammatory pain models in rats, pain measures are reduced. In this study we selectively knockout (KO) the MR in sensory neurons to determine the role of MR in sensory neurons of the mouse DRG in pain measures as MR antagonism during the local inflammation of the DRG (LID) pain model. We found that MR antagonism using eplerenone reduced evoked mechanical hypersensitivity during LID, but MR KO in paw-innervating sensory neurons only did not. This could be a result of differences between prolonged (MR KO) versus acute (drug) MR block or an indicator that non-neuronal cells in the DRG are driving the effect of MR antagonists. MR KO unmyelinated C neurons are more excitable under normal and inflamed conditions, while MR KO does not affect excitability of myelinated A cells. MR KO in sensory neurons causes a reduction in overall GR mRNA but is protective against reduction of the anti-inflammatory GRα isoform during LID. These effects of MR KO in sensory neurons expanded our understanding of MR's functional role in different neuronal subtypes (A and C neurons), and its interactions with the GR.


Asunto(s)
Dolor de la Región Lumbar , Antagonistas de Receptores de Mineralocorticoides , Ratas , Ratones , Humanos , Animales , Antagonistas de Receptores de Mineralocorticoides/farmacología , Antagonistas de Receptores de Mineralocorticoides/uso terapéutico , Receptores de Mineralocorticoides , Ratas Sprague-Dawley , Células Receptoras Sensoriales , Ganglios Espinales , Inflamación/tratamiento farmacológico
2.
Brain Behav Immun ; 113: 401-414, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37557960

RESUMEN

Satellite glial cells (SGCs) are among the most abundant non-neuronal cells in dorsal root ganglia (DRGs) and closely envelop sensory neurons that detect painful stimuli. However, little is still known about their homeostatic activities and their contribution to pain. Using single-cell RNA sequencing (scRNA-seq), we were able to obtain a unique transcriptional profile for SGCs. We found enriched expression of the tissue inhibitor metalloproteinase 3 (TIMP3) and other metalloproteinases in SGCs. Small interfering RNA and neutralizing antibody experiments revealed that TIMP3 modulates somatosensory stimuli. TIMP3 expression decreased after paclitaxel treatment, and its rescue by delivery of a recombinant TIMP3 protein reversed and prevented paclitaxel-induced pain. We also established that paclitaxel directly impacts metalloproteinase signaling in cultured SGCs, which may be used to identify potential new treatments for pain. Therefore, our results reveal a metalloproteinase signaling pathway in SGCs for proper processing of somatosensory stimuli and potential discovery of novel pain treatments.


Asunto(s)
Ganglios Espinales , Neuroglía , Humanos , Ganglios Espinales/metabolismo , Neuroglía/metabolismo , Dolor/metabolismo , Transducción de Señal , Células Receptoras Sensoriales , Análisis de la Célula Individual
3.
Expert Opin Ther Targets ; 27(8): 665-678, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37574713

RESUMEN

INTRODUCTION: Current treatments for chronic pain are inadequate. Here, we provide an update on the new therapeutic strategies that target dorsal root ganglia (DRGs) in the peripheral nervous system for a better and safer treatment of chronic pain. AREAS COVERED: Despite the complex nature of chronic pain and its underlying mechanisms, we do know that changes in the plasticity and modality of neurons in DRGs play a pivotal role. DRG neurons are heterogenous and offer potential pain targets for different therapeutic interventions. We discuss the last advancements of these interventions, which include the use of systemic and local administrations, selective nerve drug delivery, and gene therapy. In particular, we provide updates and further details on the molecular characterization of primary sensory neurons, new analgesics entering the market, and future gene therapy approaches. EXPERT OPINION: DRGs and primary sensory neurons are promising targets for chronic pain treatment due to their key role in pain signaling, unique anatomical location, and the potential for different targeted therapeutic interventions.


Asunto(s)
Dolor Crónico , Humanos , Dolor Crónico/tratamiento farmacológico , Ganglios Espinales , Analgésicos , Células Receptoras Sensoriales
4.
Neurosci Bull ; 39(9): 1363-1374, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37165177

RESUMEN

Although sympathetic blockade is clinically used to treat pain, the underlying mechanisms remain unclear. We developed a localized microsympathectomy (mSYMPX), by cutting the grey rami entering the spinal nerves near the rodent lumbar dorsal root ganglia (DRG). In a chemotherapy-induced peripheral neuropathy model, mSYMPX attenuated pain behaviors via DRG macrophages and the anti-inflammatory actions of transforming growth factor-ß (TGF-ß) and its receptor TGF-ßR1. Here, we examined the role of TGF-ß in sympathetic-mediated radiculopathy produced by local inflammation of the DRG (LID). Mice showed mechanical hypersensitivity and transcriptional and protein upregulation of TGF-ß1 and TGF-ßR1 three days after LID. Microsympathectomy prevented mechanical hypersensitivity and further upregulated Tgfb1 and Tgfbr1. Intrathecal delivery of TGF-ß1 rapidly relieved the LID-induced mechanical hypersensitivity, and TGF-ßR1 antagonists rapidly unmasked the mechanical hypersensitivity after LID+mSYMPX. In situ hybridization showed that Tgfb1 was largely expressed in DRG macrophages, and Tgfbr1 in neurons. We suggest that TGF-ß signaling is a general underlying mechanism of local sympathetic blockade.


Asunto(s)
Radiculopatía , Factor de Crecimiento Transformador beta , Ratones , Animales , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Hiperalgesia/metabolismo , Radiculopatía/tratamiento farmacológico , Radiculopatía/metabolismo , Dolor/metabolismo , Analgésicos/farmacología , Ganglios Espinales/metabolismo
5.
Pain ; 163(1): e106-e120, 2022 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-33941753

RESUMEN

ABSTRACT: Peripheral nerve regeneration is associated with pain in several preclinical models of neuropathic pain. Some neuropathic pain conditions and preclinical neuropathic pain behaviors are improved by sympathetic blockade. In this study, we examined the effect of a localized "microsympathectomy," ie, cutting the gray rami containing sympathetic postganglionic axons where they enter the L4 and L5 spinal nerves, which is more analogous to clinically used sympathetic blockade compared with chemical or surgical sympathectomy. We also examined manipulations of CCL2 (monocyte chemoattractant protein 1), a key player in both regeneration and pain. We used rat tibial nerve crush as a neuropathic pain model in which peripheral nerve regeneration can occur successfully. CCL2 in the sensory ganglia was increased by tibial nerve crush and reduced by microsympathectomy. Microsympathectomy and localized siRNA-mediated knockdown of CCL2 in the lumbar dorsal root ganglion had very similar effects: partial improvement of mechanical hypersensitivity and guarding behavior, reduction of regeneration markers growth-associated protein 43 and activating transcription factor 3, and reduction of macrophage density in the sensory ganglia and regenerating nerve. Microsympathectomy reduced functional regeneration as measured by myelinated action potential propagation through the injury site and denervation-induced atrophy of the tibial-innervated gastrocnemius muscle at day 10. Microsympathectomy plus CCL2 knockdown had behavioral effects similar to microsympathectomy alone. The results show that local sympathetic effects on neuropathic pain may be mediated in a large part by the effects on expression of CCL2, which in turn regulates the regeneration process.


Asunto(s)
Ganglios Espinales , Neuralgia , Animales , Quimiocina CCL2 , Hiperalgesia , Monocitos , Regeneración Nerviosa , Ratas , Ratas Sprague-Dawley , Simpatectomía , Nervio Tibial
6.
Neuron ; 110(2): 209-220.e6, 2022 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-34752775

RESUMEN

Spontaneous pain refers to pain occurring without external stimuli. It is a primary complaint in chronic pain conditions and remains difficult to treat. Moreover, the mechanisms underlying spontaneous pain remain poorly understood. Here we employed in vivo imaging of dorsal root ganglion (DRG) neurons and discovered a distinct form of abnormal spontaneous activity following peripheral nerve injury: clusters of adjacent DRG neurons firing synchronously and sporadically. The level of cluster firing correlated directly with nerve injury-induced spontaneous pain behaviors. Furthermore, we demonstrated that cluster firing is triggered by activity of sympathetic nerves, which sprout into DRGs after injury, and identified norepinephrine as a key neurotransmitter mediating this unique firing. Chemogenetic and pharmacological manipulations of sympathetic activity and norepinephrine receptors suggest that they are necessary and sufficient for DRG cluster firing and spontaneous pain behavior. Therefore, blocking sympathetically mediated cluster firing may be a new paradigm for treating spontaneous pain.


Asunto(s)
Ganglios Espinales , Nervios Espinales , Ganglios Espinales/fisiología , Humanos , Dolor , Células Receptoras Sensoriales , Nervios Espinales/lesiones , Sistema Nervioso Simpático/fisiología
7.
Brain Behav Immun ; 91: 556-567, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33197543

RESUMEN

Chronic low back pain is a common condition, with high societal costs and often ineffectual treatments. Communication between macrophages/monocytes (MØ) and sensory neurons has been implicated in various preclinical pain models. However, few studies have examined specific MØ subsets, although distinct subtypes may play opposing roles. This study used a model of low back pain/radiculopathy involving direct local inflammation of the dorsal root ganglia (DRG). Reporter mice were employed that had distinct fluorescent labels for two key MØ subsets: CCR2-expressing (infiltrating pro-inflammatory) MØ, and CX3CR1-expressing (resident) macrophages. We observed that local DRG inflammation induced pain behaviors in mice, including guarding behavior and mechanical hypersensitivity, similar to the previously described rat model. The increase in MØ in the inflamed DRG was dominated by increases in CCR2+ MØ, which persisted for at least 14 days. The primary endogenous ligand for CCR2, CCL2, was upregulated in inflamed DRG. Three different experimental manipulations that reduced the CCR2+ MØ influx also reduced pain behaviors: global CCR2 knockout; systemic injection of INCB3344 (specific CCR2 blocker); and intravenous injection of liposomal clodronate. The latter two treatments when applied around the time of DRG inflammation reduced CCR2+ but not CX3CR1+ MØ in the DRG. Together these experiments suggest a key role for the CCR2/CCL2 system in establishing the pain state in this model of inflammatory low back pain and radiculopathy. Intravenous clodronate given after pain was established had the opposite effect on pain behaviors, suggesting the role of macrophages or their susceptibility to clodronate may change with time.


Asunto(s)
Dolor de la Región Lumbar , Radiculopatía , Receptores CCR2 , Animales , Quimiocina CCL2 , Ácido Clodrónico , Modelos Animales de Enfermedad , Ganglios Espinales , Macrófagos , Ratones , Receptores CCR2/genética
8.
Pain ; 161(8): 1925-1936, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32701850

RESUMEN

Previous studies have shown that the peripheral nerve regeneration process is linked to pain in several neuropathic pain models. Other studies show that sympathetic blockade may relieve pain in some pain models and clinical conditions. This study examined reduction in peripheral nerve regeneration as one possible mechanism for relief of neuropathic pain by sympathetic blockade. A "microsympathectomy," consisting of cutting the gray rami containing sympathetic postganglionic axons where they enter the L4 and L5 spinal nerves, reduced mechanical hypersensitivity in 2 different rat neuropathic pain models. In the spinal nerve ligation model, in which some functional regeneration and reinnervation of the ligated spinal nerve can be observed, microsympathectomy reduced functional and anatomical measures of regeneration as well as expression of growth-associated protein 43 (GAP43), a regeneration-related protein. In the spared nerve injury model, in which functional reinnervation is not possible and the futile regeneration process results in formation of a neuroma, microsympathectomy reduced neuroma formation and GAP43 expression. In both models, microsympathectomy reduced macrophage density in the sensory ganglia and peripheral nerve. This corroborates previous work showing that sympathetic nerves may locally affect immune function. The results further highlight the challenge of improving pain in neuropathic conditions without inhibiting peripheral nerve regeneration that might otherwise be possible and desired.


Asunto(s)
Regeneración Nerviosa/fisiología , Neuralgia/fisiopatología , Traumatismos de los Nervios Periféricos , Nervios Espinales/lesiones , Nervios Espinales/fisiopatología , Simpatectomía , Animales , Modelos Animales de Enfermedad , Femenino , Ganglios Espinales/patología , Ganglios Espinales/fisiopatología , Masculino , Nervios Periféricos , Ratas , Ratas Sprague-Dawley , Nervios Espinales/patología
9.
Anesthesiology ; 132(6): 1540-1553, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32404819

RESUMEN

BACKGROUND: Patients undergoing cancer treatment often experience chemotherapy-induced neuropathic pain at their extremities, for which there is no U.S. Food and Drug Administration-approved drug. The authors hypothesized that local sympathetic blockade, which is used in the clinic to treat various pain conditions, can also be effective to treat chemotherapy-induced neuropathic pain. METHODS: A local sympathectomy (i.e., cutting the ipsilateral gray rami entering the spinal nerves near the L3 and L4 dorsal root ganglia) was performed in mice receiving intraperitoneal injections every other day of the chemotherapeutic drug paclitaxel. Sympathectomy effects were then assessed in chemotherapy-induced pain-like behaviors (i.e., mechanical and cold allodynia) and neuroimmune and electrophysiologic responses. RESULTS: Local microsympathectomy produced a fast recovery from mechanical allodynia (mean ± SD: sympathectomy vs. sham at day 5, 1.07 ± 0.34 g vs. 0.51 ± 0.17g, n = 5, P = 0.030 in male mice, and 1.08 ± 0.28 g vs. 0.62 ± 0.16 g, n = 5, P = 0.036 in female mice) and prevented the development of cold allodynia in both male and female mice after paclitaxel. Mechanistically, microsympathectomy induced transcriptional increases in dorsal root ganglia of macrophage markers and anti-inflammatory cytokines, such as the transforming growth factor-ß. Accordingly, depletion of monocytes/macrophages and blockade of transforming growth factor-ß signaling reversed the relief of mechanical allodynia by microsympathectomy. In particular, exogenous transforming growth factor-ß was sufficient to relieve mechanical allodynia after paclitaxel (transforming growth factor-ß 100 ng/site vs. vehicle at 3 h, 1.21 ± 0.34g vs. 0.53 ± 0.14 g, n = 5, P = 0.001 in male mice), and transforming growth factor-ß signaling regulated neuronal activity in dorsal root ganglia. CONCLUSIONS: Local sympathetic nerves control the progression of immune responses in dorsal root ganglia and pain-like behaviors in mice after paclitaxel, raising the possibility that clinical strategies already in use for local sympathetic blockade may also offer an effective treatment for patients experiencing chemotherapy-induced neuropathic pain.


Asunto(s)
Hiperalgesia/inducido químicamente , Hiperalgesia/prevención & control , Inflamación/inducido químicamente , Inflamación/prevención & control , Paclitaxel/efectos adversos , Simpatectomía , Animales , Antineoplásicos Fitogénicos/efectos adversos , Frío , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones
10.
Anesth Analg ; 131(1): 298-306, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31990732

RESUMEN

BACKGROUND: Anti-inflammatory corticosteroids are a common treatment for different conditions involving chronic pain and inflammation. Clinically used steroids target the glucocorticoid receptor (GR) for its anti-inflammatory effects. We previously reported that GR in sensory neurons may play central roles in some pain models and that GR immunoreactivity signal in dorsal root ganglia (DRG) decreased after local inflammation of the DRG (a model of low back pain). In the current study, we aimed to determine if similar changes in GR signal also exist in a skin inflammation model, the complete Freund's adjuvant (CFA) model (a model of peripheral inflammatory pain), in which the terminals of the sensory neurons rather than the somata are inflamed. METHODS: A low dose of CFA was injected into the hind paw to establish the peripheral inflammation model in Sprague-Dawley rats of both sexes, as confirmed by measurements of behavior and paw swelling. Immunohistochemical and western blotting techniques were used to determine the expression pattern of the GR in the inflamed hind paw and the DRGs. Plasma corticosterone levels were measured with radioimmunoassay. RESULTS: The immunohistochemical staining revealed that GR is widely expressed in the normal DRG and skin tissues. Paw injection with CFA caused upregulation of the GR in the skin tissue on postinjection day 1, mostly detected in the dermis area. However, paw inflammation significantly reduced the GR signal in the L5 DRG 1 day after the injection. The GR downregulation was still evident 14 days after CFA inflammation. On day 1, western blotting confirmed this downregulation and showed that it could also be observed in the contralateral L5 DRG, as well as in the L2 DRG (a level which does not innervate the paw). Plasma corticosterone levels were elevated in both sexes on day 14 after CFA compared to day 1, suggesting autologous downregulation of the GR by corticosterone may have contributed to the downregulation observed on day 14 but not day 1. CONCLUSIONS: There are distinctive patterns of GR activation under different pain conditions, depending on the anatomical location. The observed downregulation of the GR in sensory neurons may have a significant impact on the use of steroids as treatment in these conditions and on the regulatory effects of endogenous glucocorticoids.


Asunto(s)
Modelos Animales de Enfermedad , Hiperalgesia/metabolismo , Receptores de Glucocorticoides/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Femenino , Adyuvante de Freund/toxicidad , Hiperalgesia/inducido químicamente , Inflamación/inducido químicamente , Inflamación/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Células Receptoras Sensoriales/efectos de los fármacos
11.
Neuroscience ; 402: 51-65, 2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30699332

RESUMEN

Low back pain is a common cause of chronic pain and disability. It is modeled in rodents by chronically compressing the lumbar dorsal root ganglia (DRG) with small metal rods, resulting in ipsilateral mechanical and cold hypersensitivity, and hyperexcitability of sensory neurons. Sodium channels are implicated in this hyperexcitability, but the responsible isoforms are unknown. In this study, we used siRNA-mediated knockdown of the pore-forming NaV1.6 and regulatory NaVß4 sodium channel isoforms that have been previously implicated in a different model of low back pain caused by locally inflaming the L5 DRG. Knockdown of either subunit markedly reduced spontaneous pain and mechanical and cold hypersensitivity induced by DRG compression, and reduced spontaneous activity and hyperexcitability of sensory neurons with action potentials <1.5 msec (predominately cells with myelinated axons, based on conduction velocities measured in a subset of cells) 4 days after DRG compression. These results were similar to those previously obtained in the DRG inflammation model and some neuropathic pain models, in which sensory neurons other than nociceptors seem to play key roles. The cytokine profiles induced by DRG compression and DRG inflammation were also very similar, with upregulation of several type 1 pro-inflammatory cytokines and downregulation of type 2 anti-inflammatory cytokines. Surprisingly, the cytokine profile was largely unaffected by NaVß4 knockdown in either model. The NaV1.6 channel, and the NaVß4 subunit that can regulate NaV1.6 to enhance repetitive firing, play key roles in both models of low back pain; targeting the abnormal spontaneous activity they generate may have therapeutic value.


Asunto(s)
Ganglios Espinales/metabolismo , Dolor de la Región Lumbar/metabolismo , Canal de Sodio Activado por Voltaje NAV1.6/metabolismo , Neuronas/fisiología , Subunidad beta-4 de Canal de Sodio Activado por Voltaje/metabolismo , Potenciales de Acción , Animales , Femenino , Ganglios Espinales/lesiones , Ganglios Espinales/fisiopatología , Inflamación/metabolismo , Mediadores de Inflamación/metabolismo , Dolor de la Región Lumbar/fisiopatología , Masculino , Modelos Animales , Umbral del Dolor , Ratas Sprague-Dawley , Compresión de la Médula Espinal , Regulación hacia Arriba
12.
J Neurosci ; 39(8): 1539-1550, 2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30617209

RESUMEN

Neuropathic pain is a significant public health challenge, yet the underlying mechanisms remain poorly understood. Painful small fiber neuropathy (SFN) may be caused by gain-of-function mutations in Nav1.8, a sodium channel subtype predominantly expressed in peripheral nociceptive neurons. However, it is not clear how Nav1.8 disease mutations induce sensory neuron hyperexcitability. Here we studied two mutations in Nav1.8 associated with hypersensitive sensory neurons: G1662S reported in painful SFN; and T790A, which underlies increased pain behaviors in the Possum transgenic mouse strain. We show that, in male DRG neurons, these mutations, which impair inactivation, significantly increase TTX-resistant resurgent sodium currents mediated by Nav1.8. The G1662S mutation doubled resurgent currents, and the T790A mutation increased them fourfold. These unusual currents are typically evoked during the repolarization phase of action potentials. We show that the T790A mutation greatly enhances DRG neuron excitability by reducing current threshold and increasing firing frequency. Interestingly, the mutation endows DRG neurons with multiple early afterdepolarizations and leads to substantial prolongation of action potential duration. In DRG neurons, siRNA knockdown of sodium channel ß4 subunits fails to significantly alter T790A current density but reduces TTX-resistant resurgent currents by 56%. Furthermore, DRG neurons expressing T790A channels exhibited reduced excitability with fewer early afterdepolarizations and narrower action potentials after ß4 knockdown. Together, our data demonstrate that open-channel block of TTX-resistant currents, enhanced by gain-of-function mutations in Nav1.8, can make major contributions to the hyperexcitability of nociceptive neurons, likely leading to altered sensory phenotypes including neuropathic pain in SFN.SIGNIFICANCE STATEMENT This work demonstrates that two disease mutations in the voltage-gated sodium channel Nav1.8 that induce nociceptor hyperexcitability increase resurgent currents. Nav1.8 is crucial for pain sensations. Because resurgent currents are evoked during action potential repolarization, they can be crucial regulators of action potential activity. Our data indicate that increased Nav1.8 resurgent currents in DRG neurons greatly prolong action potential duration and enhance repetitive firing. We propose that Nav1.8 open-channel block is a major factor in Nav1.8-associated pain mechanisms and that targeting the molecular mechanism underlying these unique resurgent currents represents a novel therapeutic target for the treatment of aberrant pain sensations.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.8/fisiología , Neuralgia/fisiopatología , Nocicepción/fisiología , Enfermedades del Sistema Nervioso Periférico/fisiopatología , Células Receptoras Sensoriales/fisiología , Sodio/metabolismo , Potenciales de Acción , Animales , Modelos Animales de Enfermedad , Mutación con Ganancia de Función , Humanos , Activación del Canal Iónico , Transporte Iónico , Masculino , Ratones , Ratones Mutantes Neurológicos , Ratones Transgénicos , Mutación Missense , Canal de Sodio Activado por Voltaje NAV1.8/genética , Neuralgia/etiología , Técnicas de Placa-Clamp , Enfermedades del Sistema Nervioso Periférico/complicaciones , Mutación Puntual , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes , Células Receptoras Sensoriales/metabolismo , Tetrodotoxina/farmacología
13.
Front Cell Neurosci ; 12: 453, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30524245

RESUMEN

Low back pain, a leading cause of disability, is commonly treated by epidural steroid injections that target the anti-inflammatory glucocorticoid receptor (GR). However, their efficacy has been controversial. All currently used epidural steroids also activate the pro-inflammatory mineralocorticoid receptor (MR) with significant potency. Local inflammation of the dorsal root ganglia (DRG), a rat model of low back pain, was used. This model causes static and dynamic mechanical allodynia, cold allodynia and guarding behavior (a measure of spontaneous pain), and activates the MR, with pro-nociceptive effects. In this study, effects of local Dexamethasone (DEX; a glucocorticoid used in epidural injections), and eplerenone (EPL; a second generation, more selective MR antagonist) applied to the DRG at the time of inflammation were examined. Mechanical and spontaneous pain behaviors were more effectively reduced by the combination of DEX and EPL than by either alone. The combination of steroids was particularly more effective than DEX alone or the model alone (3-fold improvement for mechanical allodynia) at later times (day 14). Immunohistochemical analysis of the GR in the DRG showed that the receptor was expressed in neurons of all size classes, and in non-neuronal cells including satellite glia. The GR immunoreactivity was downregulated by DRG inflammation (48%) starting on day 1, consistent with the reduction of GR (57%) observed by Western blot, when compared to control animals. On day 14, the combination of DEX and EPL resulted in rescue of GR immunoreactivity that was not seen with DEX alone, and was more effective in reducing a marker for satellite glia activation/neuroinflammation. The results suggest that EPL may enhance the effectiveness of clinically used epidural steroid injections, in part by enhancing the availability of the GR. Thus, the glucocorticoid-mineralocorticoid interactions may limit the effectiveness of epidural steroids through the regulation of the GR in the DRG.

16.
Pain ; 159(9): 1731-1741, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29708941

RESUMEN

Obesity is often associated with increased pain, but little is known about the effects of obesity and diet on postoperative pain. In this study, effects of diet and obesity were examined in the paw incision model, a preclinical model of postoperative pain. Long-Evans rats were fed high-fat diet (40% calories from butter fat) or low-fat normal chow. Male rats fed high-fat diet starting 6 weeks before incision (a diet previously shown to induce markers of obesity) had prolonged mechanical hypersensitivity and an overall increase in spontaneous pain in response to paw incision, compared with normal chow controls. Diet effects in females were minor. Removing high-fat diet for 2 weeks before incision reversed the diet effects on pain behaviors, although this was not enough time to reverse high-fat diet-induced weight gain. A shorter (1 week) exposure to high-fat diet before incision also increased pain behaviors in males, albeit to a lesser degree. The 6-week high-fat diet increased macrophage density as examined immunohistochemically in lumbar dorsal root ganglion even before paw incision, especially in males, and sensitized responses of peritoneal macrophages to lipopolysaccharide stimuli in vitro. The nerve regeneration marker growth-associated protein 43 (GAP43) in skin near the incision (day 4) was higher in the high-fat diet group, and wound healing was delayed. In summary, high-fat diet increased postoperative pain particularly in males, but some diet effects did not depend on weight gain. Even short-term dietary manipulations, that do not affect obesity, may enhance postoperative pain.


Asunto(s)
Conducta Animal/fisiología , Dieta Alta en Grasa/efectos adversos , Inflamación/etiología , Dolor Postoperatorio/etiología , Animales , Ingestión de Energía/fisiología , Femenino , Proteína GAP-43/metabolismo , Inflamación/metabolismo , Macrófagos Peritoneales/metabolismo , Masculino , Dolor Postoperatorio/metabolismo , Ratas , Ratas Long-Evans , Factores Sexuales , Cicatrización de Heridas/fisiología
17.
Neurosci Bull ; 34(1): 22-41, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29333591

RESUMEN

The voltage-gated Na+ channel subtype Nav1.7 is important for pain and itch in rodents and humans. We previously showed that a Nav1.7-targeting monoclonal antibody (SVmab) reduces Na+ currents and pain and itch responses in mice. Here, we investigated whether recombinant SVmab (rSVmab) binds to and blocks Nav1.7 similar to SVmab. ELISA tests revealed that SVmab was capable of binding to Nav1.7-expressing HEK293 cells, mouse DRG neurons, human nerve tissue, and the voltage-sensor domain II of Nav1.7. In contrast, rSVmab showed no or weak binding to Nav1.7 in these tests. Patch-clamp recordings showed that SVmab, but not rSVmab, markedly inhibited Na+ currents in Nav1.7-expressing HEK293 cells. Notably, electrical field stimulation increased the blocking activity of SVmab and rSVmab in Nav1.7-expressing HEK293 cells. SVmab was more effective than rSVmab in inhibiting paclitaxel-induced mechanical allodynia. SVmab also bound to human DRG neurons and inhibited their Na+ currents. Finally, potential reasons for the differential efficacy of SVmab and rSVmab and future directions are discussed.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Canal de Sodio Activado por Voltaje NAV1.7/inmunología , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Neuralgia/tratamiento farmacológico , Neuralgia/metabolismo , Animales , Biotina/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Ganglios Espinales/citología , Células HEK293 , Humanos , Hibridomas/química , Hiperalgesia/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos C57BL , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Canal de Sodio Activado por Voltaje NAV1.7/química , Unión Proteica/efectos de los fármacos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/uso terapéutico , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/fisiología
18.
Anesth Analg ; 126(1): 150-160, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28742774

RESUMEN

BACKGROUND: Intraoperative lung-protective ventilation (ILPV) is defined as tidal volumes <8 mL/kg ideal bodyweight and is increasingly a standard of care for major abdominal surgical procedures performed under general anesthesia. In this study, we report the result of a quality improvement initiative targeted at improving adherence to ILPV guidelines in a large academic teaching hospital. METHODS: We performed a time-series study to determine whether anesthesia provider adherence to ILPV was affected by certain improvement interventions and patient ideal body weight (IBW). Tidal volume data were collected at 3 different time points for 191 abdominal surgical cases from June 2014 through April 2015. Improvement interventions during that period included education at departmental grand rounds, creation of a departmental ILPV policy, feedback of tidal volume and failure rate data at grand rounds sessions, and reducing default ventilator settings for tidal volume. Mean tidal volume per kilogram of ideal body weight (VT/kg IBW) and rates of noncompliance with ILPV were analyzed before and after the interventions. A survey was administered to assess provider attitudes after implementation of improvement interventions. Responses before and after interventions and between physician and nonphysician providers were analyzed. RESULTS: Reductions in mean VT/kg IBW and rates of failure for providers to use ILPV occurred after improvement interventions. Patients with IBW <65 kg received higher VT/kg IBW and had higher rates of failure to use ILPV than patients with IBW >65 kg. Surveyed providers demonstrated stronger agreement to having knowledge and practice consistent with ILPV after interventions. CONCLUSIONS: Our interventions improved anesthesia provider adherence to low tidal volume ILPV. IBW was found to be an important factor related to provider adherence to ILPV. Provider attitudes about their knowledge and practice consistent with ILPV also changed with our interventions.


Asunto(s)
Centros Médicos Académicos/normas , Adhesión a Directriz/normas , Pulmón/fisiología , Monitoreo Intraoperatorio/normas , Ventilación Pulmonar/fisiología , Respiración Artificial/normas , Adulto , Anciano , Femenino , Humanos , Análisis de Series de Tiempo Interrumpido/métodos , Análisis de Series de Tiempo Interrumpido/normas , Masculino , Persona de Mediana Edad , Monitoreo Intraoperatorio/métodos , Respiración Artificial/métodos , Estudios Retrospectivos , Volumen de Ventilación Pulmonar/fisiología
19.
Neurosci Bull ; 34(1): 85-97, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28534262

RESUMEN

Injury to peripheral nerves can lead to neuropathic pain, along with well-studied effects on sensory neurons, including hyperexcitability, abnormal spontaneous activity, and neuroinflammation in the sensory ganglia. Neuropathic pain can be enhanced by sympathetic activity. Peripheral nerve injury may also damage sympathetic axons or expose them to an inflammatory environment. In this study, we examined the lumbar sympathetic ganglion responses to two rat pain models: ligation of the L5 spinal nerve, and local inflammation of the L5 dorsal root ganglion (DRG), which does not involve axotomy. Both models resulted in neuroinflammatory changes in the sympathetic ganglia, as indicated by macrophage responses, satellite glia activation, and increased numbers of T cells, along with very modest increases in sympathetic neuron excitability (but not spontaneous activity) measured in ex vivo recordings. The spinal nerve ligation model generally caused larger responses than DRG inflammation. Plasticity of the sympathetic system should be recognized in studies of sympathetic effects on pain.


Asunto(s)
Ganglios Simpáticos/patología , Inflamación Neurogénica/etiología , Dolor/etiología , Dolor/patología , Traumatismos de los Nervios Periféricos/complicaciones , Potenciales de Acción/fisiología , Animales , Modelos Animales de Enfermedad , Femenino , Proteína Ácida Fibrilar de la Glía/metabolismo , Hiperalgesia/etiología , Ligadura/efectos adversos , Macrófagos/patología , Masculino , Técnicas de Placa-Clamp , Ratas , Ratas Sprague-Dawley , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo
20.
Sci Rep ; 7(1): 10350, 2017 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-28871134

RESUMEN

Obesity is associated with increased risk for chronic pain. Basic mechanisms for this association are poorly understood. Using a milder version of a radicular pain model, local inflammation of the dorsal root ganglion (DRG), we observed marked increases in mechanical and cold allodynia in rats of both sexes that were maintained on a high-fat diet (HFD) for 6 weeks prior to DRG inflammation. Notably, this increase in pain-related behaviors occurred in both Long-Evans and Sprague-Dawley rats despite the fact that the 6-week HFD exposure induced obesity (e.g., increased insulin, leptin, weight, and percent body fat) in the Long-Evans, but not Sprague-Dawley, strains. This suggested that HFD, rather than obesity per se, increased pain behaviors. Increased pain behaviors were observed even after a much shorter (1 week) exposure to the HFD but the effect was smaller. HFD also increased behavioral responses and paw swelling to paw injection of complete Freund's adjuvant, a model of peripheral inflammatory pain. No change was detected in plasma cytokine levels in HFD rats. However, increased macrophage infiltration of the DRG was observed in response to the HFD, absent any pain model. The results suggest that HFD can increase pain even when it does not cause obesity.


Asunto(s)
Conducta Animal , Dieta Alta en Grasa , Obesidad/complicaciones , Dolor/etiología , Adipoquinas/sangre , Adipoquinas/metabolismo , Animales , Biomarcadores , Peso Corporal , Citocinas/sangre , Citocinas/metabolismo , Modelos Animales de Enfermedad , Adyuvante de Freund/efectos adversos , Adyuvante de Freund/inmunología , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Inmunohistoquímica , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Dolor/diagnóstico , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA