Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Biol Rep ; 50(10): 8639-8651, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37535245

RESUMO

Extracellular vesicles (EVs) theranostic potential is under intense investigation. There is a wealth of information highlighting the role that EVs and the secretome play in disease and how these are being utilized for clinical trials and novel therapeutic possibilities. However, understanding of the physiological and pathological roles of EVs remain incomplete. The challenge lies in reaching a consensus concerning standardized quality-controlled isolation, storage, and sample preparation parameters. Interest in circulating EV cargo as diagnostic and prognostic biomarkers is steadily growing. Though promising, various limitations need to be addressed before there can be successful, full-scale therapeutic use of approved EVs. These limitations include obtaining or manufacturing from the appropriate medium (e.g., from bodily fluid or cell culture), loading and isolating EVs, stability, and storage, standardization of processing, and determining potency. This review highlights specific topics, including circulation of abnormal EVs contribute to human disease and the theranostic potential of EVs. Theranostics is defined as a combination of the word's therapeutics and diagnostics and describes how a specific medicine or technique can function as both. Key findings include, (1) EVs and the secretome are future theranostics which will be utilized as both biomarkers for diagnosis and as therapeutics, (2) basic and translational research supports clinical trials utilizing EVs/secretome, and (3) additional investigation is required to fully unmask the theranostic potential of EVs/secretome in specific diseases and injuries.


Assuntos
Vesículas Extracelulares , Humanos , Biomarcadores , Medicina de Precisão , Comunicação Celular , Técnicas de Cultura de Células
2.
Eur J Pharmacol ; 955: 175901, 2023 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-37451423

RESUMO

Neuropathic pain is caused by the lesion or disease of the somatosensory system and can be initiated and/or maintained by both central and peripheral mechanisms. Nerve injury leads to neuronal damage and apoptosis associated with the release of an array of pathogen- or damage-associated molecular patterns to activate inflammasomes. The activation of the NLR family pyrin domain containing 3 (NLRP3) inflammasome contributes to neuropathic pain and may represent a novel target for pain therapeutic development. In the current review, we provide an up-to-date summary of the recent findings on the involvement of NLRP3 inflammasome in modulating neuropathic pain development and maintenance, focusing on peripheral neuropathic conditions. Here we provide a detailed review of the mechanisms whereby NLRP3 inflammasomes contribute to neuropathic pain via (1) neuroinflammation, (2) apoptosis, (3) pyroptosis, (4) proinflammatory cytokine release, (5) mitochondrial dysfunction, and (6) oxidative stress. We then present the current research literature reporting on the antinociceptive effects of several natural products and pharmacological interventions that target activation, expression, and/or regulation of NLRP3 inflammasome. Furthermore, we emphasize the effects of microRNAs as another regulator of NLRP3 inflammasome. In conclusion, we summarize the possible caveats and future perspectives that might provide successful therapeutic approaches against NLRP3 inflammasome for treating or preventing neuropathic pain conditions.

3.
J Chem Neuroanat ; 124: 102123, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35738454

RESUMO

Preclinical and clinical studies have reported sex differences in pain and analgesia. These differences may be linked to anatomical structures of the central nervous system pain modulatory circuitry, and/or hormonal milieu. The midbrain periaqueductal gray (PAG) is a critical brain region for descending inhibition of pain. The PAG projects to the rostral ventromedial medulla (RVM), which projects bilaterally to the spinal cord to inhibit pain. In addition to pain, this descending circuit (or pathway) can be engaged by endogenous opioids (i.e., endorphins) or exogenous opioids (i.e., morphine), and we have previously reported sex differences in the activation of this circuit during pain and analgesia. Forebrain structures, including the amygdala, project to and engage the PAG-RVM circuit during persistent inflammatory pain. However, there are limited studies in females detailing this amygdalar-PAG pathway and its involvement during persistent inflammatory pain. The objective of the present study was to delineate the neural projections from the amygdala to the PAG in male and female rats to determine if they are sexually distinct in their anatomical organization. We also examined the activation of this pathway by inflammatory pain and the co-localization of receptors for estrogen. Injection of the retrograde tracer fluorogold (FG) into the ventrolateral PAG (vlPAG) resulted in dense retrograde labeling in both the central amygdala (CeA) and medial amygdala (MeA). While the number of CeA-vlPAG neurons were comparable between the sexes, there were more MeA-vlPAG neurons in females. Inflammatory pain resulted in greater activation of the amygdala in males; however, females displayed higher Fos expression within CeA-vlPAG projection neurons. Females expressed higher ERα in the MeA and CeA and the same was true of the projection neurons. Together, these data indicate that although the MeA-vlPAG projections are denser in females, inflammatory pain does not significantly activate these projections. In contrast, inflammatory pain resulted in a greater activation of the CeA-vlPAG pathway in females. As females experience a greater number of chronic pain syndromes, the CeA-vlPAG pathway may play a facilitatory (and not inhibitory) role in pain modulation.


Assuntos
Substância Cinzenta Periaquedutal , Caracteres Sexuais , Animais , Feminino , Masculino , Bulbo/metabolismo , Dor/metabolismo , Substância Cinzenta Periaquedutal/metabolismo , Ratos , Ratos Sprague-Dawley
4.
Antioxidants (Basel) ; 11(2)2022 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-35204312

RESUMO

Oxidative stress, resulting from an imbalance between the formation of damaging free radicals and availability of protective antioxidants, can contribute to peripheral neuropathic pain conditions. Reactive oxygen and nitrogen species, as well as products of the mitochondrial metabolism such as superoxide anions, hydrogen peroxide, and hydroxyl radicals, are common free radicals. Nuclear factor erythroid 2 (NFE2)-related factor 2 (Nrf2) is a transcription factor encoded by the NFE2L2 gene and is a member of the cap 'n' collar subfamily of basic region leucine zipper transcription factors. Under normal physiological conditions, Nrf2 remains bound to Kelch-like ECH-associated protein 1 in the cytoplasm that ultimately leads to proteasomal degradation. During peripheral neuropathy, Nrf2 can translocate to the nucleus, where it heterodimerizes with muscle aponeurosis fibromatosis proteins and binds to antioxidant response elements (AREs). It is becoming increasingly clear that the Nrf2 interaction with ARE leads to the transcription of several antioxidative enzymes that can ameliorate neuropathy and neuropathic pain in rodent models. Current evidence indicates that the antinociceptive effects of Nrf2 occur via reducing oxidative stress, neuroinflammation, and mitochondrial dysfunction. Here, we will summarize the preclinical evidence supporting the role of Nrf2 signaling pathways and Nrf2 inducers in alleviating peripheral neuropathic pain.

5.
Mol Pain ; 18: 17448069211069255, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35040378

RESUMO

Orofacial pain disorders are predominately experienced by women. Progesterone, a major ovarian hormone, is neuroprotective and antinociceptive. We recently reported that progesterone attenuates estrogen-exacerbated orofacial pain behaviors, yet it remains unclear what anatomical substrate underlies progesterone's activity in the trigeminal system. Progesterone has been reported to exert protective effects through actions at intracellular progesterone receptors (iPR), membrane-progesterone receptors (mPR), or sigma 1 receptors (Sig-1R). Of these, the iPR and Sig-1R have been reported to have a role in pain. Progesterone can also have antinociceptive effects through its metabolite, allopregnanolone. Two enzymes, 5α-reductase and 3α-hydroxysteroid dehydrogenase (3α-HSD), are required for the metabolism of progesterone to allopregnanolone. Both progesterone and allopregnanolone rapidly attenuate pain sensitivity, implicating action of either progesterone at Sig-1R and/or conversion to allopregnanolone which targets GABAA receptors. In the present study, we investigated whether Sig-1 Rs are expressed in nociceptors within the trigeminal ganglia of cycling female rats and whether the two enzymes required for progesterone metabolism to allopregnanolone, 5α-reductase and 3α-hydroxysteroid dehydrogenase, are also present. Adult female rats from each stage of the estrous cycle were rapidly decapitated and the trigeminal ganglia collected. Trigeminal ganglia were processed by either fluorescent immunochemistry or western blotting to for visualization and quantification of Sig-1R, 5α-reductase, and 3α-hydroxysteroid dehydrogenase. Here we report that Sig-1Rs and both enzymes involved in progesterone metabolism are highly expressed in a variety of nociceptive sensory neuron populations in the female rat trigeminal ganglia at similar levels across the four stages of the estrous cycle. These data indicate that trigeminal sensory neurons are an anatomical substrate for the reported antinociceptive activity of progesterone via Sig-1R and/or conversion to allopregnanolone.


Assuntos
Nociceptores , Progesterona , Analgésicos , Animais , Feminino , Humanos , Nociceptividade , Nociceptores/metabolismo , Progesterona/metabolismo , Progesterona/farmacologia , Ratos , Receptores sigma , Células Receptoras Sensoriais/metabolismo , Gânglio Trigeminal/metabolismo , Receptor Sigma-1
6.
Neurobiol Pain ; 10: 100073, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34504982

RESUMO

Orofacial pain disorders involving trigeminal sensory neurons disproportionately affect women and can be modulated by hormones, especially estrogen (E2). Proinflammatory mediators, like serotonin (5HT), can act on sensory neurons expressing the transient receptor potential vanilloid 1 (TRPV1) ion channel, resulting in peripheral sensitization. We previously reported peripheral 5HT evokes greater pain behaviors in the hindpaw of female rats during proestrus and estrus, stages when E2 fluctuates. It is unknown if this interaction is comparable in the trigeminal system. We hypothesized that E2 exacerbates 5HT-evoked nocifensive pain behaviors and pain signaling in female trigeminal sensory neurons. We report 5HT-evoked nocifensive behaviors are significantly higher during estrus and proestrus, which is attenuated by blocking the 5HT2A receptor. The comparable dose of 5HT was not nociceptive in males unless capsaicin was also administered. When administered with capsaicin, a lower dose of 5HT evoked trigeminal pain behaviors in females during proestrus. Further, basal 5HT content in the vibrissal pad was higher in cycling females compared to males. Ex vivo, E2 enhanced 5HT-potentiated CGRP release from trigeminal neurons, which was not significantly reduced by blocking the 5HT2A receptor. Our data indicates that estrogen fluctuation influences the pronociceptive effects of 5HT on trigeminal sensory neurons.

7.
Tex J Micros ; 52(1): 8-13, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-37309469

RESUMO

Serotonin (5-hydroxytryptamine, 5HT) is a neurotransmitter and proinflammatory mediator found largely in the peripheral nervous system where it can initiate pain signaling. 5HT binds a variety of 5HT receptors on sensory nerve endings specialized in detecting noxious stimuli, termed nociceptors. A subset of sensory neurons involved in pain signaling express the transient receptor potential vanilloid 1 ion channel (TRPV1), a pain generator. 5HT can both directly activate sensory neurons and sensitize TRPV1 leading to enhanced nociceptor sensitivity (peripheral sensitization). Previous studies in male rats reported that the 5HT receptor subtype 3A (5HT3A) and TRPV1 are co-expressed on sensory neurons, but it is unknown if 5HT3A and TRPV1 are co-expressed on female sensory neurons. Given that craniofacial pain disorders occur at a 2-3x greater prevalence in women, examining pain mechanisms in female trigeminal sensory neurons that innervate the craniofacial region is critical to advancing craniofacial pain management in women. Here we examined whether (1) 5HT acting via the 5HT3A receptor produces sexually dimorphic orofacial pain behaviors in rats and (2) whether 5HT3A receptor mRNA is expressed in trigeminal sensory neurons, including the TRPV1-expressing subpopulation, and increase pain signaling. We report that 5HT evokes pain behaviors in male and female rats, which was not significantly reduced by antagonism of 5HT3A. We performed in situ hybridization to label 5HT3A and TRPV1 mRNA in trigeminal sensory neurons and found distinct cell populations with either 5HT3A mRNA or TRPV1 mRNA in males and females. Further, 5HT3A antagonism failed to reduce pain signaling in cultured trigeminal sensory neurons. These data suggest that the 5HT3A subtype on trigeminal sensory neurons innervating the orofacial soft tissues does not play a significant role in sexually dimorphic craniofacial pain disorders. As previous studies have reported that granisetron reduces masseter muscle pain, 5HT3 may play a role in sex differences in myofascial pain disorders but not in other craniofacial pain disorders.

8.
Oxid Med Cell Longev ; 2019: 8594375, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31612077

RESUMO

Recent studies have reported that the transient receptor potential V1 ion channel (TRPV1), a pain generator on sensory neurons, is activated and potentiated by NADPH oxidase-generated reactive oxygen species (ROS). ROS are increased by advanced oxidation protein products (AOPPs), which activate NADPH oxidase by upregulating Nox4 expression. Our previous studies reported that Euphorbia bicolor (Euphorbiaceae) latex extract induced peripheral analgesia, partly via TRPV1, in hindpaw-inflamed male and female rats. The present study reports that E. bicolor latex extract also can evoke analgesia via reduction of oxidative stress biomarkers and proinflammatory cytokines/chemokines in a rat model of orofacial pain. Male and female rats were injected with complete Freund's adjuvant (CFA) into the left vibrissal pad to induce orofacial inflammation, and mechanical allodynia was measured by the von Frey method. Twenty-four hours later, rats received one injection of E. bicolor latex extract or vehicle into the inflamed vibrissal pad. Mechanical sensitivity was reassessed at 1, 6, 24, and/or 72 hours. Trigeminal ganglia and trunk blood were collected at each time point. In the trigeminal ganglia, ROS were quantified using 2',7'-dichlorodihydrofluorescein diacetate dye, Nox4 protein was quantified by Western blots, and cytokines/chemokines were quantified using a cytokine array. AOPPs were quantified in trunk blood using a spectrophotometric assay. E. bicolor latex extract significantly reduced orofacial mechanical allodynia in male and female rats at 24 and 72 hours, respectively. ROS, Nox4, and proinflammatory cytokines/chemokines were significantly reduced in the trigeminal ganglia, and plasma AOPP was significantly reduced in the trunk blood of extract-treated compared to vehicle-treated rats. In vitro assays indicate that E. bicolor latex extract possessed antioxidant activities by scavenging free radicals. Together our data indicate that the phytochemicals in E. bicolor latex may serve as novel therapeutics for treating oxidative stress-induced pain conditions.


Assuntos
Citocinas/metabolismo , Euphorbiaceae/química , Dor Facial/tratamento farmacológico , Látex/uso terapêutico , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/uso terapêutico , Animais , Modelos Animais de Doenças , Feminino , Masculino , Ratos , Ratos Sprague-Dawley
9.
Front Pharmacol ; 10: 958, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31551772

RESUMO

The negative side effects of opioid-based narcotics underscore the search for alternative non-opioid bioactive compounds that act on the peripheral nervous system to avoid central nervous system-mediated side effects. The transient receptor potential V1 ion channel (TRPV1) is a peripheral pain generator activated and sensitized by heat, capsaicin, and a variety of endogenous ligands. TRPV1 contributes to peripheral sensitization and hyperalgesia, in part, via triggering the release of proinflammatory peptides, such as calcitonin gene-related peptide (CGRP), both locally and at the dorsal horn of the spinal cord. Ultrapotent exogenous TRPV1 agonists, such as resiniferatoxin identified in the latex of the exotic Euphorbia resinifera, trigger hyperalgesia followed by long lasting, peripheral analgesia. The present study reports on the analgesic properties of Euphorbia bicolor, a relative of E. resinifera, native to the Southern United States. The study hypothesized that E. bicolor latex extract induces long-lasting, non-opioid peripheral analgesia in a rat model of inflammatory pain. Both inflamed and non-inflamed adult male and female rats were injected with the methanolic extract of E. bicolor latex into the hindpaw and changes in pain behaviors were reassessed at various time points up to 4 weeks. Primary sensory neuron cultures also were treated with the latex extract or vehicle for 15 min followed by stimulation with the TRPV1 agonist capsaicin. Results showed that E. bicolor latex extract evoked significant pain behaviors in both male and female rats at 20 min post-injection and lasting around 1-2 h. At 6 h post-injection, analgesia was observed in male rats that lasted up to 4 weeks, whereas in females the onset of analgesia was delayed to 72 h post-injection. In sensory neurons, latex extract significantly reduced capsaicin-evoked CGRP release. Blocking TRPV1, but not opioid receptors, attenuated the onset of analgesia and capsaicin-induced CGRP release. Latex was analyzed by mass spectrometry and eleven candidate compounds were identified and reported here. These findings indicate that phytochemicals in the E. bicolor latex induce hyperalgesia followed by peripheral, non-opioid analgesia in both male and female rats, which occurs in part via TRPV1 and may provide novel, non-opioid peripheral analgesics that warrant further examination.

10.
Neuropsychopharmacology ; 44(1): 155-165, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29973654

RESUMO

Morphine remains one of the most widely prescribed opioids for alleviation of persistent and/or severe pain; however, multiple preclinical and clinical studies report that morphine is less efficacious in females compared to males. Morphine primarily binds to the mu opioid receptor, a prototypical G-protein coupled receptor densely localized in the midbrain periaqueductal gray. Anatomical and physiological studies conducted in the 1960s identified the periaqueductal gray, and its descending projections to the rostral ventromedial medulla and spinal cord, as an essential descending inhibitory circuit mediating opioid-based analgesia. Remarkably, the majority of studies published over the following 30 years were conducted in males with the implicit assumption that the anatomical and physiological characteristics of this descending inhibitory circuit were comparable in females; not surprisingly, this is not the case. Several factors have since been identified as contributing to the dimorphic effects of opioids, including sex differences in the neuroanatomical and neurophysiological characteristics of the descending inhibitory circuit and its modulation by gonadal steroids. Recent data also implicate sex differences in opioid metabolism and neuroimmune signaling as additional contributing factors. Here we cohesively present these lines of evidence demonstrating a neural basis for sex differences in opioid modulation of pain, with a focus on the PAG as a sexually dimorphic core of descending opioid-induced inhibition and argue for the development of sex-specific pain therapeutics.


Assuntos
Analgésicos Opioides/uso terapêutico , Morfina/uso terapêutico , Neuroglia/metabolismo , Neurônios/metabolismo , Dor/metabolismo , Substância Cinzenta Periaquedutal/metabolismo , Caracteres Sexuais , Analgésicos Opioides/farmacologia , Animais , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Morfina/farmacologia , Neuroglia/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Dor/tratamento farmacológico , Substância Cinzenta Periaquedutal/efeitos dos fármacos , Resultado do Tratamento
11.
Neuroscience ; 384: 87-100, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29800719

RESUMO

Many persistent pain conditions occur predominantly in women making pain a major women's health issue. One theory for the prevalence in females is hormone modulation of pain mechanisms. The peripheral release of the neurotransmitter serotonin (5HT) has been implicated in various sexually dimorphic pain conditions; yet no studies have examined the effect of ovarian hormones on peripheral 5HT-evoked pain behaviors. We hypothesized that peripheral 5HT evokes greater pain behaviors in female rodents during estrus and/or proestrus, stages of the estrous cycle where ovarian hormones are greatly fluctuating. Female Sprague-Dawley rats (250-350 g) from each stage of the estrous cycle, ovariectomized females, and intact males received an intraplantar hindpaw injection of 5HT (2 µg/100 µL) or saline (n = 6 per group) and thermal hyperalgesia, mechanical allodynia, or edema was measured at 0, 10, 20 and 30 min post-injection. A separate group of rats received an ipsilateral injection of the selective 5HT2A antagonist, M100907, 15 min prior to 5HT injection. We report that females in proestrus and estrus exhibited significantly greater and/or longer lasting pain behaviors compared to males, females in diestrus, and ovariectomized females. There were no significant sex differences or estrous cycle effects on 5HT-evoked edema or 5HT content in inflamed hindpaws. Local pretreatment with the 5HT2A receptor antagonist blocked 5HT-evoked thermal hyperalgesia and edema. These data provide evidence of a modulatory role of hormones on peripheral 5HT-evoked pain occurring via the 5HT2A receptor.


Assuntos
Comportamento Animal/efeitos dos fármacos , Ciclo Estral/fisiologia , Hiperalgesia/fisiopatologia , Dor/induzido quimicamente , Serotonina/farmacologia , Caracteres Sexuais , Animais , Comportamento Animal/fisiologia , Feminino , Masculino , Ovariectomia , Dor/fisiopatologia , Medição da Dor , Ratos , Ratos Sprague-Dawley
12.
Pain Med ; 18(12): 2453-2465, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27794548

RESUMO

OBJECTIVE: Opioid-based analgesics are a major component of the lengthy pain management of burn patients, including military service members, but are problematic due to central nervous system-mediated side effects. Peripheral analgesia via targeted ablation of nociceptive nerve endings that express the transient receptor potential vanilloid channel 1 (TRPV1) may provide an improved approach. We hypothesized that local injection of the TRPV1 agonist resiniferatoxin (RTX) would produce long-lasting analgesia in a rat model of pain associated with burn injury. METHODS: Baseline sensitivities to thermal and mechanical stimuli were measured in male and female Sprague-Dawley rats. Under anesthesia, a 100 °C metal probe was placed on the right hind paw for 30 seconds, and sensitivity was reassessed 72 hours following injury. Rats received RTX (0.25 µg/100 µL; ipl) into the injured hind paw, and sensitivity was reassessed across three weeks. Tissues were collected from a separate group of rats at 24 hours and/or one week post-RTX for pathological analyses of the injured hind paw, dorsal spinal cord c-Fos, and primary afferent neuropeptide immunoreactivity. RESULTS: Local RTX reversed burn pain behaviors within 24 hours, which lasted through recovery at three weeks. At one week following RTX, decreased c-Fos and primary afferent neuropeptide immunoreactivities were observed in the dorsal horn, while plantar burn pathology was unaltered. CONCLUSIONS: These results indicate that local RTX induces long-lasting analgesia in a rat model of pain associated with burn. While opioids are undesirable in trauma patients due to side effects, RTX may provide valuable long-term, nonopioid analgesia for burn patients.


Assuntos
Analgésicos/farmacologia , Queimaduras/complicações , Diterpenos/farmacologia , Manejo da Dor/métodos , Animais , Modelos Animais de Doenças , Feminino , Masculino , Ratos , Ratos Sprague-Dawley , Canais de Cátion TRPV/agonistas
13.
Burns ; 41(8): 1796-1804, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26432505

RESUMO

Thermal burns among individuals working in highly stressful environments, such as firefighters and military Service Members, are common. Evidence suggests that pre-injury stress may exaggerate pain following thermal injury; however current animal models of burn have not evaluated the potential influence of pre-burn stress. This sham-controlled study evaluated the influence of prior stress exposure on post-burn thermal and mechanical sensitivity in male Sprague-Dawley rats. Rats were exposed to 20 min of inescapable swim stress or sham stress once per day for three days. Exposure to inescapable swim stress (1) increased the intensity and duration of thermal hyperalgesia after subsequent burn and (2) accelerated the onset of thermal hyperalgesia and mechanical allodynia after subsequent burn. This stress-induced exacerbation of pain sensitivity was reversed by pretreatment and concurrent treatment with the serotonin-norepinephrine reuptake inhibitor (SNRI) duloxetine. These data suggest a better understanding of mechanisms by which prior stress augments pain after thermal burn may lead to improved pain treatments for burn survivors.


Assuntos
Comportamento Animal/efeitos dos fármacos , Queimaduras/psicologia , Hiperalgesia/psicologia , Limiar da Dor/efeitos dos fármacos , Dor/psicologia , Estresse Psicológico/psicologia , Animais , Modelos Animais de Doenças , Cloridrato de Duloxetina/farmacologia , Masculino , Limiar da Dor/psicologia , Ratos , Ratos Sprague-Dawley , Inibidores da Recaptação de Serotonina e Norepinefrina/farmacologia
14.
BMC Anesthesiol ; 15: 135, 2015 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-26444970

RESUMO

BACKGROUND: Neuropathic pain is evasive to treat once developed, however evidence suggests that local administration of anesthetics near the time of injury reduces the development of neuropathic pain. As abnormal electrical signaling in the damaged nerve contributes to the initiation and maintenance of neuropathic pain, local administration of anesthetics prior to injury may reduce its development. We hypothesized that local treatment with bupivacaine prior to nerve injury in a rat model of spinal nerve ligation (SNL) would attenuate the initiation and/or maintenance of neuropathic pain behaviors. METHODS: On the day prior to SNL, baseline measures of pre-injury mechanical, thermal, and/or cold sensitivity were recorded in adult male Sprague-Dawley rats. Immediately prior to SNL or sham treatment, the right L5 nerve was perineurally bathed in either 0.05 mL bupivacaine (0.5 %) or sterile saline (0.9 %) for 30 min. Mechanical allodynia, thermal hyperalgesia, and/or cold allodynia were then examined at 3, 7, 10, 14 and 21 days following SNL. RESULTS: Rats exhibited both mechanical and cold allodynia, but not thermal hyperalgesia, within 3 days and up to 21 days post-SNL. No significant pain behaviors were observed in sham controls. Preemptive local bupivacaine significantly attenuated both mechanical and cold allodynia as early as 10 days following SNL compared to saline controls and were not significantly different from sham controls. CONCLUSIONS: These data indicate that local treatment with bupivacaine prior to surgical manipulations that are known to cause nerve damage may protect against the maintenance of chronic neuropathic pain.


Assuntos
Anestésicos Locais/administração & dosagem , Bupivacaína/administração & dosagem , Hiperalgesia/tratamento farmacológico , Nervos Espinhais/efeitos dos fármacos , Nervos Espinhais/lesões , Animais , Temperatura Baixa/efeitos adversos , Hiperalgesia/patologia , Ligadura , Masculino , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...